27.12.2012 Views

IN THIS ISSUE - Drug Development & Delivery

IN THIS ISSUE - Drug Development & Delivery

IN THIS ISSUE - Drug Development & Delivery

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

July/August 2012 Vol 12 No 6 www.drug-dev.com<br />

The science & business of drug development in specialty pharma, biotechnology, and drug delivery<br />

Philip Graham,<br />

PhD<br />

Deuterium<br />

Modification as a New<br />

Branch of Medicinal<br />

Chemistry to Develop<br />

Novel, Highly<br />

Differentiated <strong>Drug</strong>s<br />

Cindy H. Dubin<br />

Transdermal,<br />

Topical &<br />

Subcutaneous:<br />

Non-Invasive<br />

<strong>Delivery</strong> to Expand<br />

Product Line<br />

Extensions<br />

Jean Sung,<br />

PhD<br />

A Next-Generation<br />

Inhaled Dry Powder<br />

<strong>Delivery</strong> Platform<br />

<strong>IN</strong> <strong>THIS</strong><br />

<strong>ISSUE</strong><br />

<strong>IN</strong>TERVIEW WITH<br />

CAISSON BIOTECH’S<br />

CEO<br />

THOMAS HARLAN<br />

Regulatory<br />

Harmony 18<br />

Gill King, MBA<br />

Joel Finkle<br />

Medicated<br />

Chewing Gum 20<br />

Shivang Chaudhary,<br />

MSPharm<br />

3E<br />

Trilogy 24<br />

David F. Scelba<br />

Addressing<br />

Solubility 26<br />

Rod Ray, PhD, PE<br />

Microneedle<br />

Technology 30<br />

Mikolaj Milewski<br />

Amitava Mitra<br />

Injection<br />

Molding 36<br />

Andrew Loxley, PhD<br />

Designing<br />

Autoinjectors 41<br />

Jonathan Wilkins, PhD<br />

Iain Simpson, PhD


July/August 2012 Vol 12 No 6<br />

PUBLISHER/PRESIDENT<br />

Ralph Vitaro<br />

rvitaro@drug-dev.com<br />

EXECUTIVE EDITORIAL DIRECTOR<br />

Dan Marino, MSc<br />

dmarino@drug-dev.com<br />

CREATIVE DIRECTOR<br />

Shalamar Q. Eagel<br />

CONTROLLER<br />

Debbie Carrillo<br />

CONTRIBUT<strong>IN</strong>G EDITORS<br />

Cindy H. Dubin<br />

John A. Bermingham<br />

Josef Bossart, PhD<br />

Katheryn Symank<br />

TECHNICAL OPERATIONS<br />

Mark Newland<br />

EDITORIAL SUPPORT<br />

Nicholas D. Vitaro<br />

ADM<strong>IN</strong>ISTRATIVE SUPPORT<br />

Kathleen Kenny<br />

Corporate/Editorial Office<br />

219 Changebridge Road, Montville, NJ 07045<br />

Tel: (973)299-1200<br />

Fax: (973) 299-7937<br />

www.drug-dev.com<br />

International<br />

Ralph Vitaro<br />

219 Changebridge Road<br />

Montville, NJ 07045<br />

Tel: (973) 299-1200<br />

Fax: (973) 299-7937<br />

E-mail: rvitaro@drug-dev.com<br />

Mailing List Rental<br />

Candy Brecht<br />

Tel: (703) 706-0383<br />

Fax: (703) 549-6057<br />

E-mail: cbrecht@mgilists.com<br />

Advertising Sales Offices<br />

West Coast<br />

Warren De Graff<br />

818 5th Avenue, Suite 301<br />

San Rafael, CA 94901<br />

Tel: (415) 721-0644<br />

Fax: (415) 721-0665<br />

E-mail: wjdegraff@drug-dev.com<br />

All editorial submissions are handled with reasonable care, but the publishers assume no responsibility for the safety<br />

of artwork, photographs, or manuscripts. Every precaution is taken to ensure accuracy, but publishers cannot accept<br />

responsibility for the accuracy of information supplied herein or for any opinion expressed. <strong>Drug</strong> <strong>Development</strong> &<br />

<strong>Delivery</strong> (ISSN) 1537-2898 is published 9 times in 2012, January/February, March, April, May, June, July/August,<br />

September, October, and November/December by <strong>Drug</strong> <strong>Delivery</strong> Technology LLC, 219 Changebridge Road, Montville NJ<br />

07045. Subscription rates: $120.00 for 1 year in the United States, Canada, and Mexico. $188.00 for 1 year outside<br />

the United States, Canada, and Mexico. All subscriptions are payable in US funds, drawn on US banks. Send payment<br />

to: <strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> LLC subscription Department, 219 Changebridge Road, Montville NJ 07045. Single<br />

copies (prepaid) $20.00, US, Canada, and Mexico; $25.00 in all other countries. Add $5.00 per order for shipping<br />

and handling. Periodicals Postage Paid at Montville, NJ 07045-9998 and additional mailing offices. Postmaster:<br />

please send address changes to <strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong>, 219 Changebridge Road, Montville NJ 07045. All rights<br />

reserved under the US International and Pan-American Copyright Conventions. All rights reserved. No part of this<br />

publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including by<br />

photocopy, recording, or information storage and retrieval system, without written permission from the publisher.<br />

Authorization to photocopy items for internal or personal use, or the internal or personal use of specific clients, is<br />

granted by <strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> for libraries and other users registered with the Copywrite Clearance, 222<br />

Rosewood Drive, Danvers, MA 01923; phone: (978) 750-8400, fax: (978) 750-4470.<br />

Proud member of<br />

East & Midwest<br />

Patricia Loyas<br />

977 Wall St.<br />

Diamond Point, NY 12824<br />

Tel: (518) 668-4144<br />

Fax: (518) 668-9794<br />

E-mail: ployas@drug-dev.com


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

6<br />

Addressing<br />

Solubility<br />

“With BCS Class II compounds comprising<br />

an increasing percentage of compounds in<br />

our clients' discovery portfolios, Bend<br />

Research is positioned to continue to<br />

provide the highest-quality support and<br />

innovation for our clients' pipelines. This<br />

proactive support is focused on two crucial<br />

areas: (1) support of the existing<br />

technology platforms and (2) development<br />

of next-generation technologies.”<br />

p.26<br />

24 3E Trilogy: Entertain, Educate &<br />

Engage….Become an Infotainer<br />

David F. Scelba continues his multiple part series on effective<br />

messaging and communications in the life science industry.<br />

26 Addressing Solubility Challenges:<br />

Using Effective Technology & Problem-<br />

Solving for <strong>Delivery</strong> Solutions<br />

Rod Ray, PhD, reviews his spray-dried dispersion (SDD)<br />

technology, which is recognized as a reliable solution to oral<br />

bioavailability challenges because of its proven performance,<br />

predictable long-term stability, and excellent manufacturability.<br />

30 Recent <strong>Development</strong>s in Microneedle<br />

Technology for Transdermal <strong>Drug</strong><br />

<strong>Delivery</strong> & Vaccination<br />

Mikolaj Milewski and Amitava Mitra highlight some of the recent<br />

advances in the field of microneedle-mediated transdermal drug<br />

delivery and vaccination, including summaries of preclinical<br />

pharmacokinetic data and a brief overview of clinical studies,<br />

encompassing the time period from 2010 to present day.<br />

36 Injection Molding in the<br />

Pharmaceutical Industry<br />

Andrew Loxley, PhD, and Brett Braker say many of the processes<br />

used to manufacture products within the pharmaceutical<br />

industry are unique to the particular product; however, there are<br />

also processes that have been borrowed and adapted from other<br />

manufacturing industries and successfully employed in the<br />

development of pharmaceutical products. One such example is<br />

injection molding.<br />

41 Mathematical Modelling for Faster<br />

Autoinjector Design<br />

Jonathan Wilkins, PhD, and Iain Simpson, PhD, believe a fast<br />

and effective approach to a better injection device design is a<br />

novel combination of mathematical modeling on a desktop PC,<br />

supported with complementary experimental data.<br />

46 A Next-Generation Inhaled Dry<br />

Powder <strong>Delivery</strong> Platform<br />

Jean C. Sung, PhD, says that while delivery of drugs via first-<br />

generation inhaled drug systems provides great advantages over<br />

oral or intravenous delivery, these systems also have inherent<br />

limitations, creating a tremendous opportunity for next-<br />

generation inhaled delivery platforms.


8<br />

New<br />

Medicines<br />

“Concert Pharmaceuticals, a clinical-stage<br />

biopharmaceutical company, uses deuterium to<br />

create and develop highly differentiated new<br />

medicines. This is a significant departure from<br />

the traditional use of deuterium modification<br />

as an analytical probe for metabolic studies.<br />

The company uses its DCE PlatformTM (Deuterium Chemical Entity) to develop new<br />

drugs that are designed to have first-in-class<br />

or best-in-class efficacy and/or safety.<br />

Deuterium modification provides a novel<br />

opportunity to develop new drugs by building<br />

upon existing scientific or clinical experience,<br />

potentially significantly reducing time, risk,<br />

and expense.”<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6 Developing<br />

p.67<br />

51 Transdermal, Topical & Subcutaneous:<br />

Non-Invasive <strong>Delivery</strong> to Expand<br />

Product Line Extensions<br />

Special Feature: Contributor Cindy H. Dubin asked delivery<br />

system providers and contract developers and manufacturers to<br />

describe their products and service offerings in their respective<br />

areas of expertise and how they are changing the overall<br />

landscape of the transdermal, topical, and subcutaneous markets.<br />

59 Caisson Biotech: Innovation in <strong>Drug</strong><br />

<strong>Delivery</strong> Using a Naturally Occurring<br />

Sugar Molecule<br />

<strong>Drug</strong> <strong>Development</strong> Executive: Thomas Harlan, CEO of Caisson,<br />

discusses how his company is improving the quality and delivery<br />

of numerous medications, making life easier for patients, and<br />

offering new ways for companies to enhance their drug pipeline.<br />

67 Deuterium Modification as a New<br />

Branch of Medicinal Chemistry to<br />

Develop Novel, Highly Differentiated<br />

<strong>Drug</strong>s<br />

Philip Graham, PhD; Julie Liu, PhD; and David Turnquist, MBA;<br />

show that deuterium modification may be used broadly to<br />

improve upon previously known compounds or their analogs, in<br />

turn offering potential benefits in a wide range of therapeutic<br />

areas.<br />

71 Accera, Inc: Discovering<br />

Breakthroughs in Treating Central<br />

Nervous System Disorders<br />

Executive Summary: Holger Kunze, CEO of Accera, discusses the<br />

company's novel approach to treating AD by addressing cerebral<br />

hypometabolism.<br />

DEPARTMENTS<br />

Market News & Trends . . . . . . . . . . . . . . . . . . . . . 12<br />

Regulatory Review . . . . . . . . . . . . . . . . . . . . . . . . 18<br />

A Shrinking World: Realizing Regulatory Harmony<br />

Excipient Update . . . . . . . . . . . . . . . . . . . . . . . . . 20<br />

Directly Compressible “Medicated Chewing Gum<br />

(MCG)” for Staying Alert<br />

Technology & Services Showcase . . . . . . . . . . . . 62<br />

External <strong>Delivery</strong> . . . . . . . . . . . . . . . . . . . . . . . . . 74<br />

On Behalf of Private Equity


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

10<br />

Dan Marino, MSc<br />

Executive Director<br />

<strong>Drug</strong> <strong>Development</strong><br />

& <strong>Delivery</strong><br />

Shaukat Ali, PhD,<br />

MSc<br />

Technical Service Manager<br />

BASF Pharma Solutions<br />

Sarath Chandar, MBA<br />

Vice President, Global<br />

Marketing & Commercial<br />

<strong>Development</strong><br />

SPI Pharma<br />

Ms. Debra Bingham<br />

Partner<br />

Valeo Partners<br />

Philip Green, PhD<br />

Senior Director,<br />

<strong>Drug</strong> <strong>Delivery</strong> Devices<br />

Merck Bioventures,<br />

Merck Research<br />

Laboratories<br />

James N. Czaban, JD<br />

Chairma, FDA Practice Group<br />

Wiley Rein LLP<br />

Josef Bossart, PhD<br />

Managing Director<br />

The Pharmanumbers Group<br />

Peter Hoffmann, PhD<br />

Biopharmaceutical<br />

Executive<br />

James D. Ingebrand,<br />

MBA<br />

VP & General Manager<br />

3M <strong>Drug</strong> <strong>Delivery</strong><br />

Systems<br />

The<br />

EDITORIAL<br />

Advisory Board<br />

John A.<br />

Bermingham<br />

Co-President & COO<br />

Agratech, Inc.<br />

Derek G. Hennecke<br />

President & CEO<br />

Xcelience<br />

Clifford M.<br />

Davidson, Esq.<br />

Founding Partner<br />

Davidson, Davidson &<br />

Kappel, LLC<br />

Keith Horspool, PhD<br />

Vice President,<br />

Pharmaceutics<br />

Boehringer Ingelheim<br />

Uday B. Kompella,<br />

PhD<br />

Professor, Department of<br />

Pharmaceutical Sciences<br />

University of Colorado<br />

Denver<br />

Marc Iacobucci,<br />

MBA<br />

VP, Marketing & Project<br />

Management<br />

DPT Laboratories<br />

Cornell Stamoran<br />

VP, Strategy & Corporate<br />

<strong>Development</strong><br />

Catalent Pharma<br />

Solutions<br />

Beth A-S. Brown, PhD,<br />

MS<br />

Senior Research Scientists II,<br />

Formulation & Process<br />

<strong>Development</strong><br />

Gilead Sciences<br />

Der-Yang Lee, PhD<br />

Senior Research Fellow<br />

Global Technology, R&D,<br />

McNeil Consumer Healthcare,<br />

Johnson & Johnson<br />

Gary W. Cleary, PhD,<br />

PharmD, MBA<br />

President & CTO<br />

Corium International<br />

John Fraher<br />

President<br />

Aptalis Pharmaceutical<br />

Technologies<br />

Firouz Asgarzadeh,<br />

PhD<br />

Melt Extrusion Strategic<br />

Team Lead<br />

Evonik-Degussa<br />

Corporation<br />

James W. McGinity,<br />

PhD<br />

Professor of Pharmaceutics<br />

University of Texas<br />

at Austin<br />

Wei-Guo Dai, PhD<br />

Fellow, <strong>Drug</strong> <strong>Delivery</strong> &<br />

Device <strong>Development</strong><br />

J&J Pharmaceutical R&D,<br />

LLC<br />

Matthew D. Burke, PhD<br />

Principal Scientist,<br />

Exploratory Pharmaceutics<br />

GlaxoSmithKline<br />

Robert W. Lee, PhD<br />

Vice President,<br />

Pharmaceutical<br />

<strong>Development</strong><br />

Particle Sciences


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

12<br />

GSK Successfully Uses Capsugel’s Licaps<br />

Capsugel recently announced that its Licaps liquid-filled<br />

hard capsules have been successfully used by major<br />

healthcare company, GlaxoSmithKline (GSK), for the most<br />

established brand in the German urological market, GRANU<br />

F<strong>IN</strong>K.<br />

After initially launching the product in a soft gelatin<br />

capsule form, GSK’s Consumer Healthcare Division decided to<br />

investigate other dosage form options to try and improve the<br />

product’s quality and reduce its production-to-market lead time.<br />

GRANU F<strong>IN</strong>K’s active ingredients, pumpkin seeds extracts,<br />

and pumpkin oil, were challenging to encapsulate because of<br />

the large particle size and required a long maturing period<br />

when encapsulated in traditional softgels. After testing a switch<br />

to Capsugel’s Licaps technology, GSK found the product to be<br />

leak-free and have now reduced their product lead times by an<br />

amazing 12 weeks.<br />

"After comparing two different automated capsule sealing<br />

techniques, the Licaps capsules had intact seals during the<br />

complete period of stability testing, and we could not detect<br />

any leakage,” said Dr. Stephan Wurtz, Head of Production at<br />

GSK in Herrenberg. “Capsule integrity is a key parameter used<br />

to predict product shelf-life and especially helps with<br />

preventing too many product returns, and this is why we<br />

ultimately chose the Licaps liquid-filled hard capsules as our<br />

dosage form."<br />

Since Licaps capsules entered the market nearly 10 years<br />

ago, Capsugel has made multiple technological advances,<br />

including an enhanced mechanical seal with a seal zone six<br />

times greater than a banded capsule. And while they are able to<br />

effectively mask taste and odor, Licaps capsules also offer<br />

many branding options that may improve loyalty when<br />

compared with other dosage forms. According to Capsugel’s<br />

research, consumers perceive Licaps capsules to be highquality,<br />

modern, and natural looking in a dosage form that is<br />

memorable, easy to recognize, and easy to swallow. GSK also<br />

conducted their own consumer survey to test the visual and<br />

handling acceptance of the Licaps dosage form among regular<br />

users of GRANU F<strong>IN</strong>K femina, and 73% of consumers were<br />

positive about the new Licaps liquid-filled hard capsule format.<br />

And Licaps liquid-filled hard capsules provide a solution<br />

for a variety of challenging compounds. It can improve<br />

bioavailability for poorly soluble compounds while providing<br />

effective formulation options for low-melting point compounds<br />

and safe formulation of low-dose/high potency actives. Licaps<br />

manufacturing also offers an established and robust process<br />

with the flexibility to allow for production in-house or by<br />

Capsugel. With a straightforward manufacturing transfer,<br />

including the installation of Liquid Encapsulation Microspray<br />

Sealing (LEMS) technology, Licaps capsules can even use the<br />

same line as powder-filled capsules and are sealed and dried in<br />

a matter of minutes, making manufacturing a cost-effective and<br />

environmentally friendly process.<br />

"The Licaps system has integrated well with our Bosch<br />

capsule filling equipment, and because of the expert technical<br />

assistance we had from Capsugel's installation and formulation<br />

teams, we put in place a number of new validated production<br />

processes with five fully trained operators in a very short<br />

time,” added Dr. Wurtz. “During one shift using the Licaps<br />

system, up to 350,000 Licaps capsules can be filled. We now<br />

have full in-house control of the manufacture of our drug and<br />

supplement lines, which gives us a number of benefits."


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

14<br />

Cook Pharmica Receives Milestone FDA Approval<br />

Cook Pharmica LLC recently announced it has received its<br />

first approval from the US FDA to manufacture<br />

commercial product. The product approval for commercial<br />

manufacturing came from the FDA’s Center for Biologics<br />

Evaluation and Research following a rigorous pre-approval<br />

inspection of Cook Pharmica in February. The inspection<br />

covered the company’s new vial filling line, lyophilization<br />

capabilities, and facility-wide quality systems.<br />

“This FDA decision marks a very important milestone for<br />

our company,” said Tedd Green, President of Cook Pharmica.<br />

“This first commercial manufacturing approval opens Cook<br />

Pharmica’s doors to the vibrant biopharmaceutical industry as it<br />

generates breakthroughs for patients in the United States and<br />

worldwide.”<br />

Company executives noted that the FDA’s approval of its<br />

vial filling line and lyophilization (freeze-drying) capabilities<br />

has already generated widespread industry interest in its full<br />

manufacturing and packaging capabilities, with inquiries coming<br />

from many leading biopharmaceutical companies. The rigorous<br />

preapproval inspection is a prerequisite for commercial<br />

Catalent & Bend Research Form Strategic Partnership<br />

Catalent Pharma Solutions, Inc. and Bend Research Inc.<br />

recently announced they have entered into an agreement to<br />

provide integrated solutions for pharmaceutical companies<br />

seeking to develop and manufacture specialized multiparticulate<br />

oral controlled-release products.<br />

Under the agreement, Bend Research and Catalent will<br />

provide an integrated approach to bring complex controlled-<br />

release products to market faster and more efficiently with<br />

optimal therapeutic and release profiles.<br />

The companies’ combined expertise in formulation<br />

development and Catalent's breadth of services in<br />

analytical/CMC, solid-state optimization, clinical and<br />

commercial supply will provide pharmaceutical companies with<br />

optimal dosage forms and a more efficient path to market.<br />

Catalent and Bend Research are developing joint operations and<br />

technology-transfer protocols to make the customer experience<br />

seamless and efficient while leveraging the strengths of both<br />

companies to develop better treatments for patients globally.<br />

“Our integrated approach is geared toward complex,<br />

multiparticulate controlled-release products, which traditionally<br />

have presented a high scale-up risk when they are transferred to<br />

manufacturing of clients’ drug product.<br />

“This approval demonstrates Cook Pharmica’s ability and<br />

commitment to manufacture high-quality drug products within<br />

the FDA’s stringent regulatory standards and practices. Everyone<br />

at Cook Pharmica is very pleased to have the opportunity and<br />

responsibility to ensure that we deliver a dependable commercial<br />

drug-product supply for our clients and ultimately to patients,”<br />

added Mr. Green.<br />

The parenteral manufacturing business unit of Cook<br />

Pharmica contains both vial and prefilled syringe manufacturing<br />

lines under barrier isolation, as well as secondary packaging to<br />

prepare products for distribution. Capable of filling 15 million<br />

vials and 70 million prefilled syringes annually, Cook<br />

Pharmica’s assets are available to fill the shortage of domestic<br />

drug product manufacturing in the United States and support<br />

federal efforts to eliminate dangerous drug shortages<br />

domestically. Based on other client projects at Cook Pharmica,<br />

the company expects to receive similar approvals to manufacture<br />

drug products for overseas markets as well.<br />

commercial manufacturing sites,” said Rod Ray, CEO of Bend<br />

Research. “This partnership with Catalent will provide an<br />

efficient pathway for these medicines from early development<br />

through commercialization. We believe that Catalent's breadth of<br />

services and demonstrated success in bringing controlled-release<br />

products to market, as well as supplying them globally, makes<br />

them an ideal complement to our development strengths.”<br />

“Catalent and Bend are aligning their scientific expertise<br />

and processes to ensure that developments are undertaken from<br />

Day One based on Quality by Design principles,” added Ian<br />

Muir, President of Catalent’s Modified Release Technologies<br />

business. “Bend’s added laboratory scale modeling expertise will<br />

enhance and increase the efficiencies that Catalent will provide<br />

to customers to bring difficult to formulate and manufacture<br />

controlled-release compounds to market faster, with optimal<br />

product profiles. This should enable optimal and seamless scale-<br />

up within Catalent’s Controlled Release network, and<br />

particularly at our Winchester, KY, facility, which is widely<br />

regarded as the leading commercial facility for multiparticulate<br />

products.”


Hovione Reports Significant Sales<br />

Growth<br />

Hovione recently announced that the consolidated sales<br />

for the fiscal year ended March 2012 amounted to<br />

$180 million, the sixth consecutive year of sales growth,<br />

representing a growth of 24% in relation to last year.<br />

“Another year of continued strong performance by the<br />

Hovione group. During the last 5 years, Hovione has<br />

doubled its sales and has made bold strategic steps to both<br />

strengthen its ability to serve innovators and to consolidate<br />

its leadership in off-patent contrast agents. Looking forward,<br />

and despite the difficult economic environment, we remain<br />

confident that 2012 will be another year of solid growth,”<br />

said Miguel Calado, Chief Financial Officer.<br />

In addition to the financial results, which reflect the<br />

quality of the Team’s performance, overall, 2011 represented<br />

a year of great achievements, namely Hovione stood behind<br />

three NDA approvals (these were all major NMEs) and in<br />

two cases, the approvals were full QbD filings in which<br />

Hovione was central to the design and data generation.<br />

All Hovione plants underwent several successful GMP<br />

inspections by one or more of the major Medicines’<br />

Agencies - a reflection of the large flow of filings and the<br />

high standards of compliance.<br />

“Getting multiple NDA approvals every year is<br />

becoming a habit at Hovione, which reflects well both on<br />

our customers, on our team, and on the CMO model. Our<br />

patient investment in capacity, new technologies, and<br />

development methodologies is paying off.” said Guy Villax,<br />

Chief Executive Officer.<br />

Hovione is a global company with over 50 years’<br />

experience in Active Pharmaceutical Ingredient and<br />

Intermediate <strong>Drug</strong> Product development and compliant<br />

manufacture. With four FDA-inspected sites in the US,<br />

China, Ireland, and Portugal, the company focuses on the<br />

most demanding customers, in the most regulated markets.<br />

Hovione offers integrated API, particle design, and<br />

formulation development and manufacturing. In the<br />

inhalation area, Hovione is the only independent company<br />

offering such a broad range of services.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

15


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

16<br />

AmDerma & Oculus Enter Multi-Country Agreement<br />

AmDerma Pharmaceuticals, LLC, a privately owned<br />

company (and parent company of Quinnova<br />

Pharmaceuticals, Inc.) and Oculus Innovative Sciences recently<br />

announced the execution of an agreement to develop and<br />

commercialize Oculus’ novel proprietary Microcyn Technology<br />

drug compounds for major dermatological conditions, including<br />

acne. The exclusive agreement includes licensing of the<br />

dermatology compounds in the US and India, with a first right<br />

of refusal for all member states of the European Union, Canada,<br />

Brazil, and Japan. Oculus retains all rights for the rest of the<br />

world.<br />

Under the terms of the agreement, Oculus received an<br />

undisclosed upfront payment, and will receive multiple clinically<br />

based payments upon achievement of several development and<br />

regulatory milestones for both acne and secondary indications,<br />

as well as future escalating double-digit royalties on net sales of<br />

products. The agreement also memorializes the intent for future<br />

joint development of additional dermatological products and<br />

indications.<br />

"This agreement reflects the AmDerma/Quinnova<br />

commitment to the future of dermatology. Microcyn compounds<br />

have the potential to bring about significant advancement in the<br />

treatment of skin diseases, which affect millions of patients,<br />

without adding to the growing problem of antibiotic resistance<br />

or steroid overuse," said Jeffrey Day, Quinnova President. "Our<br />

dermatology customers have been pleased with improved patient<br />

outcomes as a result of adopting the Microcyn-based<br />

compounds for treatment of atopic dermatitis and related skin<br />

diseases. We look to build upon this success by growing the<br />

family of Microcyn-based dermatology compounds for myriad<br />

skin afflictions."<br />

AmDerma will be responsible for the development costs for<br />

the acne formulation as well as other dermatological<br />

compounds.<br />

"Oculus is thrilled to expand the scope of our existing<br />

partnership with Quinnova and its parent company, AmDerma.<br />

Quinnova is doing an exceptional job with the commercial<br />

launch of the Atrapro product, and we are confident they are the<br />

right partner to commercialize this technology in acne and other<br />

dermatological indications," said Hoji Alimi, Founder and<br />

President of Oculus.


Ambrx & Merck to Design & Develop Biologic <strong>Drug</strong> Conjugates<br />

Ambrx recently announced the company has entered into a<br />

collaboration with Merck to design and develop rationally<br />

optimized biologic drug conjugates based on Ambrx's site-<br />

specific protein medicinal chemistry technology.<br />

"Ambrx's technology has the potential to provide the<br />

foundation for a new family of biologic drug conjugates that<br />

selectively deliver small molecules to their site of action," said<br />

Peter G. Schultz, PhD, a scientific founder and board member.<br />

"Merck's deep disease area expertise made it the partner of<br />

choice in expanding the application of this technology beyond<br />

oncology to other important disease areas."<br />

Under the terms of the agreement, Merck gains worldwide<br />

rights to develop and commercialize biotherapeutic drug<br />

conjugates directed toward a number of pre-specified targets.<br />

Ambrx will receive an upfront payment of $15 million and is<br />

eligible to receive milestone payments totaling up to $288<br />

million for successful discovery, development, and<br />

commercialization of candidates to all pre-specified targets. In<br />

addition, Ambrx will receive royalties on any net sales of<br />

products resulting from the collaboration.<br />

"Collaborations are an important part of our strategy to<br />

develop a portfolio of next-generation protein therapeutics that<br />

may offer significant benefits to patients," said Richard Murray,<br />

PhD, Senior Vice President and Head of Biologics and Vaccines<br />

Research at Merck. "This agreement will allow us to combine<br />

Ambrx's expertise in site-specific protein conjugation chemistry<br />

with Merck's expanding antibody capabilities and extensive<br />

small molecule resources."<br />

By combining the targeting properties of biologics with the<br />

potent therapeutic properties of small molecules, Merck and<br />

Ambrx plan to design and optimize new ways to specifically<br />

deliver pharmacologically active compounds to their site of<br />

action while minimizing the potential for systemic effects.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

17


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

18<br />

A Shrinking World: Realizing Regulatory Harmony<br />

By: Gill King, MBA, and Joel Finkle<br />

The life sciences industry faces a number of tough challenges in<br />

2012 - not the least of them the growing reach of increasingly<br />

onerous regulatory requirements.<br />

While new international market opportunities are emerging all<br />

the time, exploiting them remains challenging because of the differing<br />

ways each region and country handle regulatory submissions. Even<br />

when submissions follow the guidance of the International Conference<br />

on Harmonization (ICH) - which indeed has brought greater cohesion<br />

to the ways such activities are managed across the three major<br />

markets of the US, the European Union (EU), and Japan - there<br />

remain differences in interpretation involving submission formats,<br />

such as the electronic Common Technical Document (eCTD). To be<br />

able to take full advantage of all of the market opportunities open to<br />

them, companies must ensure their submissions can meet the<br />

requirements of the evolving formats, the developing standards, and<br />

the old and new regulations stipulated by the respective authorities.<br />

The following explores some of the major developments that<br />

have taken place throughout the past year in major markets and what<br />

those developments mean for the industry in the future.<br />

FDA SAFETY DRIVE PROMPTS MODERNIZATION<br />

Throughout the past 1 or 2 years, the US FDA has adopted an<br />

increasingly aggressive approach to ensuring patient safety, triggering<br />

a shake-up in quality control across all aspects of drug production and<br />

marketing. As part of its modernization drive, the FDA has been<br />

working collaboratively with standards-setting bodies - in particular,<br />

the Clinical Data Interchange Standards Consortium - to develop<br />

standards for the submission of study data. The efforts of such bodies<br />

have resulted in the development of numerous clinical standards, such<br />

as the Study Data Tabulation Model for representation of clinical trial<br />

tabulations, the Analysis Data Model for clinical trial analysis files,<br />

and the Standard for Exchange of Nonclinical Data for representation<br />

of nonclinical animal toxicology studies tabulations.<br />

Although adoption has been largely voluntary, the FDA, having<br />

developed standards in accordance with the industry’s requests, now<br />

expects companies to adopt the standards more consistently. That<br />

includes the way regulatory submissions get made. From 2015, all<br />

submissions will need to be filed in eCTD format. The Center for<br />

<strong>Drug</strong> Evaluation and Research (CDER) already receives 400 to 600<br />

electronic submissions per day; 70% of NDAs are eCTDs, and even<br />

<strong>IN</strong>DAs are gathering momentum, with 52% of <strong>IN</strong>Ds now filed as<br />

eCTDs. 1<br />

MODULE 1, VERSION 2<br />

Each agency participating in the ICH eCTD standard has its own<br />

requirements for the Module 1, or Regional, section. The FDA has<br />

just released draft guidance for an updated version of its Module 1<br />

specifications. For drug sponsors, two aspects of the new Module 1<br />

guidance are probably most significant: the introduction of bundled<br />

submissions and the decision to bring marketing submissions into the<br />

eCTD fold.<br />

The right to bundle submissions offers huge time and costsavings<br />

for the industry by allowing information from more than one<br />

product or regulatory activity to be sent in a single package. In the<br />

meantime, the FDA department that handles marketing submissions<br />

has been designated an office in its own right - the Office of<br />

Prescription <strong>Drug</strong> Promotion (OPDP) - and there is now greater<br />

differentiation between promotional material for healthcare<br />

professionals and that for direct-to-consumer advertising. 2<br />

Once the updated Module 1 has been implemented, the OPDP<br />

will accept eCTDs through the electronic gateway, and new headings<br />

and a altered hierarchy mean companies will have to use only one<br />

kind of software to send material to the FDA. There remain some<br />

issues to address, and Module 1 is currently under review. Final<br />

guidance is expected to be issued at the end of 2012, with<br />

implementation starting no earlier than January 2013 as part of a<br />

phased transition. 3<br />

EU TURBULENCE MIXED ECTD UPTAKE<br />

The fragile euro isn’t the only source of turmoil in Europe.<br />

Regulatory submission management behavior is similarly inconsistent.<br />

The eCTD is now well established at the European Medicines Agency,<br />

which mandated the eCTD for the Centralized Procedure as of<br />

January 2010. 4 Yet the Centralized Procedure accounts for only 11%<br />

of EU submissions. Of the remaining 89%, 12.7% are under the<br />

Mutual Recognition Procedure, 23.8% are under the National<br />

Procedure, and 61.8% are under the Decentralized Procedure.<br />

The EU has also struggled to gain much buy-in for the so-called<br />

variation guideline, implemented in 2010. In the past, no matter what<br />

change was being made to a product license, companies had to get<br />

approval first. In an attempt to minimize the administrative burden,<br />

the European Medicines Agency has permitted companies to<br />

implement variations by simply submitting an annual report to the<br />

agency within 12 months. In practice, however, the processes that<br />

have been instituted at most companies in Europe don’t allow for an<br />

automatic trigger that advises when to send something to the agency.


Further guidance covers work-sharing<br />

procedures and grouped variations, which the<br />

FDA is just now catching up to via its draft<br />

Module 1 specification’s bundled applications,<br />

but the guidance needs updating to cater to<br />

eCTD submissions. The uptake has been<br />

slower than the health agencies might have<br />

hoped a situation now compounded by the<br />

EudraVigilance Medicinal Product Dictionary<br />

(EVMPD) mandate requiring that by July 2 of<br />

this year, marketing authorization holders<br />

submit comprehensive EVMPD data for every<br />

medicinal product authorized in the EU.<br />

JAPAN & CH<strong>IN</strong>A COMMIT TO<br />

ECTD<br />

The eCTD is now well accepted by the<br />

Japanese regulatory authority, the<br />

Pharmaceuticals and Medical Devices Agency<br />

(PDMA), and to date, the agency has received<br />

106 official eCTDs. 5<br />

The PDMA updated its eCTD questionand-answer<br />

section in April 2011 and<br />

announced that companies in the industry<br />

could now e-mail questions to the agency.6 In<br />

addition, the PDMA released eCTD validator<br />

version 2.0 in February 2011, the first time<br />

the tool had been revised since its release 2<br />

years earlier. The new release streamlines<br />

processes and makes it more adaptable for<br />

companies. In July 2011, the PDMA released<br />

the fourth edition of the eCTD Creation<br />

Guide, which included not only updates by the<br />

Japanese authority but also industry best<br />

practices and experience. Companies are also<br />

better able to share information through the<br />

Japan eCTD Forum, which in October 2011,<br />

introduced an English home page. The forum<br />

is open to companies that have submitted<br />

eCTDs, and to date, all 37 companies that<br />

have submitted eCTDs to the authority have<br />

joined. 6<br />

Meanwhile, China’s State Food and <strong>Drug</strong><br />

Administration (SFDA) has taken firm steps<br />

to adopt processes that are more in line with<br />

international submission standards and has<br />

been looking at ways of implementing the<br />

eCTD. 6 While most submissions are still<br />

required to be in paper format, the agency has<br />

taken several steps toward electronic adoption.<br />

For one thing, it provides an electronic<br />

application form, which includes summaries<br />

of each of the different parts of the<br />

submission. At the evaluation stage, the SFDA<br />

requests certain elements in electronic format,<br />

including quality specifications, packaging,<br />

labeling, the manufacturing process, and the<br />

clinical databases. At the marketing phase,<br />

companies are being asked to submit<br />

electronic copies of their adverse-drugreaction<br />

reports, their quality specifications,<br />

and their ongoing package inserts.<br />

The SFDA is even working to set up a<br />

safe e-submissions gateway, similar to the<br />

FDA’s Electronic Submissions Gateway.<br />

Before it does so, however, it needs to prepare<br />

its own internal systems to handle the<br />

electronic components.<br />

All of this will go a long way to appease<br />

multinational companies that have been eager<br />

for China to adopt ICH standards. At present,<br />

companies filing in China are finding they<br />

have to reformat entire submissions, and<br />

standardization would certainly serve to assist<br />

with the submission process.<br />

SUMMARY<br />

Efforts continue to create greater global<br />

standardization around the way submissions<br />

are formatted, though there’s clearly still a<br />

long way to go. The longer-term goal of<br />

harmonizing the submission process - at least<br />

among ICH member nations, through the<br />

Regulated Product Submission (RPS) -<br />

represents an ideal to aim for. The third draft<br />

of release 2 has been approved at HL7 as a<br />

Draft Standard for Test Usage and is now at<br />

ICH for testing.<br />

If all goes according to plan, the RPS is<br />

expected to achieve a normative standard by<br />

this time next year, with adoption by the FDA<br />

a year later. The RPS working group will also<br />

work to earn International Organization for<br />

Standardization certification - one of the<br />

conditions the rest of the ICH requires for its<br />

adoption. It is hoped the EU and Japan will<br />

then adopt the standard by 2015, allowing<br />

RPS to become the first true and<br />

internationally ratified standard. u<br />

REFERENCES<br />

1. Hussong G, CDER Presentation, <strong>Drug</strong><br />

Information Association (DIA) Electronic<br />

Regulatory Submissions Meeting,<br />

November 2011.<br />

2. Kieste M, OPDP Presentation, DIA<br />

Electronic Regulatory Submissions<br />

Meeting. November 2011.<br />

3. Gray M, CDER Presentation, DIA<br />

Electronic Regulatory Submissions<br />

Meeting, November 2011.<br />

4. Wagner H-G, European Medicines Agency<br />

Presentation, DIA Electronic Regulatory<br />

Submissions Meeting, November 2011.<br />

5. eCTD in Japan, Atsushi Tomaru, Kyowa<br />

Hakko Kirin Co. Ltd., December 5, 2011.<br />

6. Section on China from December 7, 2011,<br />

discussion with Jeanie Kwon, Regulatory<br />

Operations Director, CSC Life Sciences.<br />

B I O G R A P H I E S<br />

Gillian King has<br />

worked in the<br />

pharmaceutical<br />

industry for over 12<br />

years, from<br />

establishing<br />

electronic document<br />

management systems<br />

for Europe, managing<br />

global submissions<br />

for NCEs, through to<br />

leading global<br />

functional teams. Her<br />

career has taken her into Pharmacovigilance,<br />

quality standards, and clinical operations, but her<br />

main focus is regulatory affairs and regulatory<br />

operations. Today, her role as Director, Head of<br />

Global Consulting within the Regulatory Solutions<br />

Group of CSC, allows her to immerse herself in<br />

challenging and complex business situations, and<br />

how organizational design, technology, and the<br />

regulatory environment work together to help<br />

achieve robust business operating models. Her<br />

achievements include setting up global regulatory<br />

operations groups and in establishing business<br />

processes and functional group design for global<br />

development and maintenance of products. She<br />

specializes in the “total view” and how cross<br />

functional departments must work together to<br />

achieve practical results. Ms. King earned her MBA<br />

from Warwick Business School, UK specializing in<br />

operations management and strategy. She can be<br />

reached at gking32@csc.com.<br />

Joel Finkle is<br />

Senior Strategist,<br />

Regulatory<br />

Informatics within<br />

CSC’s Regulatory<br />

Solutions Group<br />

(formerly ISI). He is<br />

the architect for two<br />

of their Document<br />

Creation solutions:<br />

ISIRender and<br />

ISIWriter. In his<br />

nearly 7 years, he has performed business process<br />

consulting, provided customizations to our<br />

solutions, and developed several software business<br />

partnerships. In his current role, he is working to<br />

find novel ways to solve regulatory software and<br />

service processes for customers, as well as<br />

providing the focal point for industry standards and<br />

regulatory guidance. Mr. Finkle comes from a<br />

background in the pharmaceutical industry, with 26<br />

years experience in software development and<br />

support of electronic submissions, publishing, and<br />

document templates, from custom CANDAs through<br />

eCTDs. He is currently a member of the HL7<br />

Regulated Product Submissions (RPS) standard<br />

development team, the DIA Electronic Regulatory<br />

Submission SIAC Core Team, the DIA Cross-SIAC<br />

EDM Reference Model development team, and the<br />

OASIS DITA Pharmaceutical <strong>Development</strong> Team. He<br />

can be reached at jfinkle@csc.com.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

19


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

20<br />

Directly Compressible “Medicated Chewing Gum<br />

(MCG)” for Staying Alert<br />

By: Shivang Chaudhary, MSPharm; Aliasgar Shahiwala, PhD; and Manju Misra, PhD, MPharm<br />

Medicated chewing gums are<br />

defined by the European<br />

Pharmacopoeia and the<br />

guidelines for pharmaceutical dosage forms<br />

as “solid single dose preparations with a<br />

base consisting mainly of gum that are<br />

intended to be chewed but not swallowed,<br />

providing a slow steady release of the<br />

medicine contained.” 1,2 They can be used for<br />

local treatment of mouth disease or<br />

systemic delivery by direct intraoral<br />

absorption through the buccal mucosa. 3<br />

Medicated chewing gums offer numerous<br />

advantages over other drug delivery<br />

systems, among which some important<br />

advantages are highlighted in this<br />

discussion.<br />

There is an increasing need to<br />

reformulate existing drugs into Novel <strong>Drug</strong><br />

<strong>Delivery</strong> Systems (NDDS) to extend or<br />

protect product patents thereby delaying,<br />

reducing, or avoiding generic erosion at<br />

patent expiry. By formulating the drugs in<br />

MCG composition, revitalization of old<br />

products and reformulation of new patented<br />

products is possible to distinguish from<br />

future generics competition in the market.<br />

Medicated Chewing Gum (MCG) is a<br />

solid single-dose preparation comprising<br />

gum base and other ingredients and<br />

containing active ingredient(s) that are<br />

released by chewing and intended for quick<br />

onset of action by direct intraoral<br />

absorption into systemic circulation. 4<br />

Traditionally, MCG has remained as a<br />

niche category due to its complex<br />

formulation and manufacturing that uses<br />

hot mixing and extrusion procedures<br />

involving specific technology and<br />

equipment that most pharmaceutical<br />

companies are not familiar with. Moreover,<br />

the use of heat and liquids in these<br />

processes prevents many APIs from being<br />

considered due to their sensitivity to high<br />

temperature levels and moisture.<br />

However, in the recent past, a new<br />

door has opened with the launch of<br />

innovative directly compressible powder<br />

excipients for production of medicated or<br />

functional chewing gums. These are<br />

designed specifically to bring gum<br />

technology closer to pharmaceutical<br />

companies by adapting to traditional oral<br />

solid formulation and production<br />

requirements in the pharma industry.<br />

They are made as an “all in one”<br />

combination of different ingredients that<br />

promote long chewing consistency because<br />

they contain the essential gum base, and at<br />

the same time, they have great flowability<br />

and compressibility properties due to their<br />

polyol and glidant content in order to be<br />

easily compressed and adapt to<br />

pharmaceutical production standards.<br />

The combination of these ready-made<br />

gum excipients allows multiple possibilities<br />

of formulation with an active ingredient<br />

and different flavors of choice, all in a dry<br />

room temperature process.<br />

In the present study, MCG was<br />

formulated using Health in Gum ® directly<br />

compressible powder (gum powder) with<br />

100 mg of caffeine. Pharmacopoeia quality<br />

control parameters were measured along<br />

with a human volunteer study to measure<br />

the API release rate from the chewing gum<br />

matrix.<br />

Caffeine is a highly soluble and highly<br />

permeable central nervous system (CNS)<br />

stimulant used in management of fatigue<br />

and increasing alertness, and from studies,<br />

it has been revealed that chewing action<br />

itself enhances 25% blood flow to the brain<br />

resulting in improvement in alertness. 2,3<br />

Thus, caffeine chewing gum can address<br />

F I G U R E 1


the issues of staying alert in two ways: one<br />

from drug (caffeine) and another from a drug<br />

delivery system (chewing gum). Therefore,<br />

caffeine chewing gum can be a synergistic<br />

delivery option for staying alert, especially for<br />

professions in which staying alert for long<br />

periods of time is necessary and vital.<br />

FORMULATION DEVELOPMENT<br />

The starting raw material of the formulation is<br />

the gum powder, which will account for at<br />

least 85% of the total weight of the tablet in<br />

order to have chewing gum consistency over<br />

time. Once the API and flavor combination<br />

was figured out in the correct proportions, the<br />

mixing process started as follows:<br />

Mixing Procedure<br />

• Mixing of liquid flavor on the powder gum<br />

preparation for 5 minutes<br />

• Screening of the mixed preparation through<br />

No. 22 sieve<br />

• Mixing of API, antisticking agent, flavoring<br />

ingredients, lubricant, and glidant<br />

• Sieving and blending (10 minutes)<br />

Finally, the formulation was compressed. In<br />

process quality control (IP-QC), parameters<br />

for optimized batch are shown in Table 1,<br />

which indicates it had very good flow<br />

property and compressibility.<br />

CFN-MCG QUALITY EVALUATION<br />

A. OFFICIAL (BP/EP)<br />

PRODUCT QUALITY<br />

ASSESSMENT TESTS<br />

An assay for content uniformity and<br />

friability test was carried out as per European<br />

Parameters<br />

pharmacopoeia. The final MCG formulation<br />

passed the test for uniformity of mass; none<br />

deviated from ± 5% of average mass of MCG.<br />

All 10 MCGs, which were sampled randomly,<br />

have passed the test for uniformity of content<br />

because contents of CFN in all 10 MCGs have<br />

fallen within compliance limit of 85% to<br />

115%, and average content of CFN was found<br />

to be 99.21% ± 0.53%. In friability testing,<br />

after 100 rotations, total weight loss of 10<br />

MCG was found to be 0.14%, which was less<br />

than the compliance limit of 1.0%. Thus the<br />

final MCG formulations passed in friability<br />

test as well.<br />

CFN-MCG performance evaluation for<br />

determining the percent of drug release was<br />

obtained by a “Chew Out” study of six<br />

volunteers in which each person chewed one<br />

sample of the caffeine chewing gum for<br />

different time periods (5, 10, 15, and 20<br />

mins). Next, residual drug had been cut into<br />

small pieces, frozen, and then ground till<br />

obtaining fine powder, and then analyzed to<br />

determine residual drug content by UV visible<br />

spectrophotometer at 273 nm.<br />

So, actual drug content minus residual<br />

drug content equals released drug content<br />

from the MCG.<br />

The drug contained within the MCG is<br />

T A B L E 1<br />

Result With Indication<br />

Angle of repose of gum powder 29.11° - Very good as per EP<br />

Carr’s compressibility index of gum<br />

powder<br />

8.00 - Very good as per EP<br />

Compatibility of caffeine with gum In DSC spectrum, separate peaks at 92°C (xylitol), 95°C<br />

powder by DSC<br />

(sorbitol), and 236°C (caffeine) - No Incompatibility<br />

Results of properties of the powder gum.<br />

T A B L E 2<br />

released in the saliva for the duration of the<br />

chewing process, and then it would be either<br />

absorbed through the oral mucosa or if<br />

swallowed, would be absorbed through the<br />

gastrointestinal tract. Pharmacokinetics can be<br />

determined from withdrawn blood samples at<br />

specific time intervals.<br />

SELECTION & OPTIMIZATION<br />

OF FACTORS AFFECT<strong>IN</strong>G %<br />

DRUG RELEASE FROM MCG<br />

BY 3 2 FULL FACTORIAL<br />

EXPERIMENTAL DESIGN<br />

Independent significant factors, chewing<br />

time (A) and amount of gum powder (B),<br />

affecting dependent factor (% CFN release<br />

from MCG) were first extracted out by means<br />

of ANOVA, and then extracted factors were<br />

optimized by 32 full factorial experimental<br />

design. Here, full factorial 32 designs were<br />

used for the optimization procedure because it<br />

is suitable for investigating the quadratic<br />

response surfaces and for constructing a<br />

second-order polynomial model, thus enabling<br />

optimization of the chewing time and amount<br />

of gum base to achieve sufficient drug release<br />

from MCG. Mathematical modeling,<br />

No. Official Tests Observations Compliance Criteria Result<br />

01 Uniformity of Mass None deviated from ± 5% NMT 2 deviate from ± 5% Pass<br />

02 Uniformity of Content<br />

All 10 MCG contain CFN within<br />

limits<br />

85% < x < 115% Pass<br />

03 Friability Testing<br />

Total weight loss = 0.14% of 10<br />

MCG<br />

Weight loss < 1.00% Pass<br />

Results of official product quality assessment tests.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

21


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

22<br />

evaluation of the ability to fit to the model,<br />

and response surface methodology (RSM)<br />

were performed by employing Design-Expert ®<br />

software (Version 7.1.2, Stat-Ease Inc.,<br />

Minneapolis, MN). RSM is a collection of<br />

mathematical and statistical techniques useful<br />

for the modeling and analysis of problems in<br />

which a response of interest is influenced by<br />

several variables and the objective is to<br />

optimize this response. The most extensive<br />

applications of RSM are in the industrial<br />

world, particularly in situations where several<br />

input variables potentially influence some<br />

performance measure or quality characteristic<br />

of the product or process. This performance<br />

measure or quality characteristic is called the<br />

response.<br />

A.<br />

B.<br />

Factors<br />

(Independent Variables)<br />

Chewing time (mins.)<br />

Amount of gum powder<br />

(%)<br />

Table 3 summarizes the independent and<br />

dependent variables along with their coded<br />

and actual levels. A total of four experimental<br />

testing runs were conducted.<br />

It was illustrated from the surface<br />

response graph that as chewing time (A)<br />

increases, percent of drug release increases as<br />

well, but an increasing amount of gum<br />

powder (B) has very little drop-off effect on<br />

T A B L E 3<br />

Results of official product quality assessment tests.<br />

Levels Used<br />

-1 0 +1<br />

5<br />

70<br />

10<br />

75<br />

T A B L E 4<br />

Experimental<br />

Variable Factors in Coded tTerms<br />

(Actual Terms)<br />

Test Run Chewing Time Amt. of Gum<br />

(mins.)<br />

Powder (%)<br />

1 -1(05) -1(70)<br />

2 0(10) -1(70)<br />

3 +1(15) -1(70)<br />

4 - 1(05) 0(75)<br />

5 0(10) 0(75)<br />

6 +1(15) 0(75)<br />

7 -1(05) +1(80)<br />

8 0(10) +1(80)<br />

9 +1(15) +1(80)<br />

Experimental testing runs with values of variable factors.<br />

15<br />

80<br />

Response<br />

(Dependent Variables)<br />

% <strong>Drug</strong> Release<br />

percent of caffeine release. Specifically: %<br />

<strong>Drug</strong> Release = + 88.33 + 5.67 A - 1.50 B +<br />

0.25 AB - 2.00 A²- 0.50 B².<br />

The quadratic models generated by<br />

regression analysis were used to construct a<br />

two-dimensional contour plot and threedimensional<br />

response surface plot in which<br />

response parameter DR was represented by a<br />

curvature surface as a function of A and B.<br />

Figure 1 shows the effect of chewing time and<br />

amount of gum powder in contour plot as well<br />

as response surface plot.<br />

A numerical optimization technique<br />

using the desirability approach was employed<br />

to develop a new formulation with the desired<br />

responses. In this study, optimization was<br />

performed with constraints for DR (90% <<br />

DR < 95%) set as goals to locate the optimum<br />

settings of the independent variables in the<br />

new formulation. The optimal parameters to<br />

achieve predicted CFN release of 92% (90%<br />

to 95%) as calculated from the predicted<br />

equation of drug release (A: chewing time =<br />

15 minutes, and B: amount of gum powder =<br />

75%).<br />

CONCLUSION<br />

Optimized formulation of directly<br />

compressed CFN-MCG has achieved the<br />

following:<br />

• Has passed all official MCG quality<br />

tests, including uniformity of mass,<br />

assay for uniformity of content, and<br />

friability testing as per compliance<br />

criteria mentioned in the official<br />

monograph of MCG in BP.<br />

• Released an average 92% of CFN<br />

(n=6) within 15 minutes of chewing,<br />

which is half of the normal average<br />

chewing time, in an in vivo chew out<br />

study.<br />

• Interindividual variability in percent<br />

CFN-release was observed in only up<br />

to 1 to 3 minutes, afterward, much less<br />

interindividual variability was<br />

observed in percent CFN release.<br />

Thus, the present study demonstrated<br />

that CFN could be successfully delivered by<br />

MCG into systemic circulation via direct<br />

intraoral buccal absorption.<br />

Concerning statistical analysis, it was<br />

shown that a 32 full factorial experimental<br />

design (FFED) and optimization technique<br />

can be successfully used in the development<br />

of an optimized formulation of MCG and for<br />

deciding appropriate chewing time for<br />

sufficient drug release. The optimized<br />

formulation exhibited drug release profiles<br />

that were close to the predicted responses,<br />

which was confirmed by high significant r2 =<br />

0.988 (> 0.9) value.<br />

Based on the overall results, it was<br />

concluded that the developed formulation of<br />

directly compressible taste-masked MCG of<br />

caffeine presents a better alternative to any<br />

other dosage form, including tea or coffee,<br />

because it is a synergistic delivery option for


staying alert. Moreover, CFN-MCG can be<br />

taken anywhere and anytime without<br />

preventing the consumer from living an active<br />

life, which promotes higher acceptance and<br />

compliance.<br />

Moreover today, it is known that chewing<br />

gum has other benefits: it promotes oral<br />

health, it is a mild stress relief, and it reduces<br />

the appetite sensation, all of which are added<br />

positive characteristics that increase the value<br />

of a caffeine drug delivery system in the form<br />

of chewing gum.This eventually can lead to<br />

the development of a new attractive market<br />

directed to energy and sports products, which<br />

is already flourishing in occidental countries.<br />

Other potential applications for MCG<br />

include areas like allergy, cough and cold,<br />

digestive, and oral care, added to the<br />

consolidated available nicotine and motion<br />

sickness gums. u<br />

REFERENCES<br />

1. European Pharmacopoeia, 2.9.5., 2.9.6,<br />

2.9.7, 2.9.25, 2.9.36, 6th edition.<br />

2. Wilson and Gisvold's Textbook of Organic<br />

Medicinal and Pharmaceutical Chemistry.<br />

11th ed. New York, NY: Lippincott<br />

Williams & Wilkins Publishing; 2004:511-<br />

512.<br />

3. Adopted from<br />

www.chewinggumfacts.com/chewing-gumbenefits/health-benefits-of-gum-chewing.<br />

4. Jacobsen, Christrup-LL. Medicated<br />

chewing Ggum: Ppros and cons. Am J<br />

<strong>Drug</strong> Del. 2004;2(4):75-88.<br />

B I O G R A P H I E S<br />

Shivang Chaudhary is a Formulation Scientist with a<br />

BPharm from Nirma University of Science & Technology,<br />

India, and an MSPharm in Pharmaceutics from the National<br />

Institute of Pharmaceutical Education & Research (NIPER),<br />

Ahmedabad. During his Masters he felt a necessity of<br />

reformulating existing drugs into Novel <strong>Drug</strong> <strong>Delivery</strong><br />

Systems (NDDS) to extend or protect product patents<br />

thereby delaying, reducing, or avoiding generic erosion at<br />

patent expiry. Apart from this chewing gum containing<br />

Caffeine (Alllert®) for staying alert, he has also developed<br />

medicated chewing gum containing different drugs for various other diseases, such as<br />

MCG for Erectile Dysfunction (EreX®) in vanilla, chocolate, strawberry, wild cherry,<br />

pineapple, and butterscotch flavors; MCG for motion sickness (Gaglet®) in cardamom,<br />

orange, lemon(lime), and ginger flavors; MCG for Vit-B12 deficiency (Smirk®) in banana<br />

flavor; and MCG for acidity (Relaaax®) in peppermint and spearmint flavors. He strongly<br />

believed that by formulating the drugs in MCG composition, revitalization of old<br />

products and reformulation of new patented products is possible to distinguish from<br />

future generics competition in the global market.<br />

Dr. Aliasgar Shahiwala is currently working as an<br />

Associate Professor in Dubai Pharmacy College. Before<br />

joining Dubai Pharmacy College, he was associated with the<br />

National Institute of Pharmaceutical Education and<br />

Research-Ahmedabad, India. Dr. Shahiwala has earned<br />

Masters and Doctorate degrees in Pharmaceutics and<br />

Pharmaceutical Technology from The Maharaja Sayajirao<br />

University of Baroda, India, and has completed 1-year of<br />

post-doctoral research experience at Northeastern University,<br />

USA. He has more than 3 years of rich research experience<br />

in the formulation and development division of large-scale manufacturers of<br />

pharmaceuticals in India and more than 7 years of teaching experience at both the<br />

graduate and post-graduate level. His research credentials have established him a<br />

reviewer, a member of editorial board, a speaker, and an invited author for various<br />

pharmaceutical scientific journals and conferences. Dr. Shahiwala’s key strength is<br />

diversity of work experience in different settings.<br />

Dr. Manju Misra is currently serving as Assistant Professor<br />

at NIPER Ahmedabad. She has prior experience working as<br />

part of an ANDA formulation development team at Macleods<br />

Pharmaceuticals, Mumbai, and Cadila Pharmaceuticals,<br />

Ahmedabad. She earned her PhD from BIT Mesra, Ranchi,<br />

India, and her MPharm from Nagpur University, India.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

23


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

24<br />

MARKET<strong>IN</strong>G MATTERS<br />

3E Trilogy: Entertain, Educate &<br />

Engage…..Become an Infotainer<br />

A multiple part series on effective messaging and communications in the life<br />

science industry.<br />

By: David F. Scelba, Partner, LifeSciencePR


In my previous column (May 2012,<br />

page 24-25), I provided a general<br />

overview of my four trilogies and<br />

four simple steps to implementing a<br />

social media strategy. In this column,<br />

I’ll explain in greater detail my 3E<br />

Trilogy …Entertain, Educate &<br />

Engage.<br />

Now, it doesn’t really matter if<br />

you’re in the Life Science industry or<br />

frankly any other marketplace. The<br />

fact is that effective communications<br />

means commanding and keeping your<br />

audience’s attention, and the most<br />

effective way to keep their attention is<br />

to entertain them.<br />

One of the best teachers I had in<br />

high school was my history teacher<br />

Mr. De Nooyer. He was someone who<br />

clearly understood the importance of<br />

entertaining to educate, and he<br />

engaged every student, even those with<br />

challenging attention spans. As an<br />

example, he would dress up in colonial<br />

garb when he was discussing the<br />

Revolutionary War. He even wore a<br />

German uniform and spoke with a<br />

German accent to dramatize the<br />

unspeakable evils of the concentration<br />

camps of World War II. At the time,<br />

we all thought he was nuts, but the<br />

truth is, he was a master<br />

communicator. His classroom antics<br />

and theater helped the lessons “come<br />

alive” in an incredibly entertaining and<br />

memorable way. It’s been more than 40<br />

years since I sat in his class, but I can<br />

still remember his lessons like it was<br />

yesterday. Mr. De Nooyer was a<br />

teacher’s teacher and an incredible<br />

salesman.<br />

With today’s technologies and<br />

multiple social media outlets, Mr. De<br />

Nooyer’s entertaining lessons could<br />

have been delivered and distributed to<br />

an infinite number of virtual<br />

classrooms and inquisitive students<br />

throughout the world.<br />

But while YouTube, Facebook,<br />

Twitter, LinkedIn, Pinterest, and all the<br />

other social media outlets deliver<br />

content to the right targeted audiences,<br />

they won’t gain an audience if the<br />

content isn’t entertaining.<br />

The key to implementing a<br />

successful social media strategy is the<br />

development of interesting content<br />

and, of course, the creative execution.<br />

You’ve all heard the saying, “Content<br />

is King,” and it’s absolutely true, but<br />

only if it’s presented in an entertaining<br />

manner.<br />

By now everyone knows video is<br />

the most efficient and effective<br />

communications tactic used<br />

throughout all websites, blogs, social<br />

media networks, and mobile apps. Just<br />

as Mr. De Nooyer was never<br />

embarrassed about wearing uniforms<br />

or acting in front of the live class, you<br />

shouldn’t be self conscious or feel silly<br />

reading from a teleprompter in front of<br />

a video camera.<br />

So let your creative juices flow<br />

and become an “Infotainer” with the<br />

ultimate goal of entertaining your<br />

audience. If you develop good content<br />

and creatively present it, you’ll educate<br />

your audience and engage them in<br />

ongoing communications. Engagement<br />

acts as one measurement matrix of<br />

your content and creativity, and is an<br />

indicator of whether you’re doing a<br />

good job communicating.<br />

Entertain, Educate & Engage …<br />

my simple 3E Trilogy to better, more<br />

effectively, and efficiently leverage<br />

social media communications to<br />

achieve your company’s strategic<br />

marketing and communications<br />

initiatives! u<br />

B I O G R A P H Y<br />

David F. Scelba is the Founder and<br />

Chairman of SGW Integrated Marketing &<br />

Communications and is a Partner at<br />

LifeSciencePR. He is responsible for the<br />

development of the company’s new<br />

interactive products and services and plays a<br />

key role as senior strategist for developing<br />

clients’ integrated marketing<br />

communications programs. He is also<br />

involved in researching and investigating<br />

acquisition opportunities and for initiating<br />

negotiations on behalf of the company. As a<br />

consultant to the broadcast, computer, and<br />

telephone industries Mr. Scelba experienced<br />

the technology convergence first hand. This<br />

unique background provides him the ability<br />

to develop innovative products and services<br />

that generate the most cost-effective and<br />

efficient marketing strategies available<br />

today. His diversified B2B, consumer, and<br />

retail experience encompasses industries<br />

such as: automotive; biochemical;<br />

broadcast; education (K-12colleges/universities);<br />

healthcare; hospitals;<br />

life science; microwave; pharmaceutical<br />

(research/drug delivery); political;<br />

professional video/audio; medical;<br />

telecommunications; and more. He is a<br />

keynote motivational speaker whose<br />

audiences include marketing professionals,<br />

college professors, MBA graduate students,<br />

and undergraduates seeking careers in the<br />

marketing- and communications-related<br />

industries. He also mentors business and<br />

government leaders on the use of<br />

technologically innovative tools for better<br />

communication with their targeted<br />

audiences. Mr. Scelba earned his BA and MA<br />

in education, a CFP Certification, with series<br />

6 qualifications, Health/Life and Real Estate<br />

Licenses, which contribute to his common<br />

sense marketing philosophy.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

25


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

26<br />

BIOAVAILABILITY<br />

E N H A N C E M E N T<br />

Addressing Solubility Challenges: Using<br />

Effective Technology & Problem-Solving<br />

for <strong>Delivery</strong> Solutions<br />

By: Rod Ray, PhD, PE<br />

SCIENCE & TECHNOLOGY<br />

TO SOLVE<br />

BIOAVAILABILITY<br />

CHALLENGES<br />

<strong>Delivery</strong> of compounds with poor<br />

aqueous solubility is a common<br />

problem as more than 50% of new<br />

chemical entities fit into Class II of<br />

the Biopharmaceutical Classification<br />

System (BCS). BCS Class II<br />

compounds are poorly soluble but<br />

highly permeable, meaning that most<br />

cannot be absorbed during normal<br />

gastrointestinal transit times without<br />

an enabling formulation.<br />

While there are many types of<br />

enabling formulations that improve the<br />

solubility of low-solubility<br />

compounds, they are generally based<br />

<strong>IN</strong>TRODUCTION<br />

Oral bioavailability represents a significant challenge to the pharmaceutical industry as more than half of the<br />

compounds in early development are considered poorly soluble. Bend Research, a problem-solving drug formulation<br />

development and manufacturing company, is well known for its spray-dried dispersion (SDD) technology, which is recognized<br />

as a reliable solution to this challenge because of its proven performance, predictable long-term stability, and excellent<br />

manufacturability.<br />

This review is a follow-up to an interview published in the May 2012 issue of <strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> in which the<br />

company’s background and problem-solving approach to drug delivery challenges faced by the pharmaceutical industry were<br />

discussed.<br />

on three approaches: (1) use of highenergy<br />

crystalline forms, such as salt<br />

forms, co-crystals, and crystals with<br />

reduced particle size; (2) dissolution<br />

of the drug in a lipid vehicle or lipid,<br />

solvent, and surfactant vehicle (to<br />

create a self-emulsifying dosage<br />

form); or (3) manufacture of an<br />

F I G U R E 1<br />

amorphous dispersion. Amorphous<br />

dispersions are generally<br />

manufactured by spray-drying or a<br />

hot-melt process. Each of these<br />

approaches is viable, depending on the<br />

compound’s physical and chemical<br />

properties, and all have been used for<br />

commercial pharmaceutical products.


CHOOS<strong>IN</strong>G THE APPROPRIATE<br />

TECHNOLOGY<br />

Bend Research takes an “agnostic”<br />

approach to choosing the appropriate<br />

solubilization technology, guided by a<br />

number of factors, including the<br />

compound’s physical-chemical properties,<br />

the projected dose, the desired release<br />

profile, and the results from numerous<br />

formulation and process models that we<br />

have developed throughout the past<br />

15 years.<br />

The goal in choosing the optimum<br />

solubilization technology is aimed at<br />

solving the right problem for that specific<br />

compound. Bend Research is capitalized<br />

for formulation development and cGMP<br />

manufacturing for spray-drying, hot-melt<br />

extrusion, and particle-size reduction to<br />

support our agnostic approach.<br />

Additionally, we can support the<br />

formulation of self-emulsifying and lipid<br />

formulations relying on a partner to<br />

complete the cGMP manufacturing.<br />

While we consider all possible<br />

technologies as we develop formulation<br />

approaches for poorly soluble compounds,<br />

our experience in the formulation of more<br />

than 500 low-solubility compounds is that<br />

spray-drying amorphous dispersions is the<br />

most widely applicable approach. This<br />

process involves dissolving the compound<br />

and a concentration-sustaining polymer in<br />

a volatile organic solvent that is rapidly<br />

removed in the spray dryer. This process<br />

allows for rapid drying of the formulation<br />

and isolation of the amorphous dispersion.<br />

It is widely applicable in that it avoids<br />

either melting the compound in a hot-melt<br />

process or relying on the solubility of the<br />

compound in a lipid vehicle.<br />

An additional benefit is that the<br />

spray-drying process has been scaled<br />

down to small scales compatible with<br />

early preclinical quantities of compound<br />

and scaled up to the multi-ton scales<br />

necessary for commercial manufacture.<br />

SPRAY-DRIED DISPERSIONS:<br />

KEY FACTORS & PROCESS<br />

OVERVIEW<br />

There are three key components to<br />

obtaining a high-performing amorphous<br />

dispersion. These are the spray-drying<br />

process conditions, the identification of<br />

the compound’s physical-chemical<br />

properties, and finally, the choice of<br />

excipients that are used in the amorphous<br />

dispersion.<br />

The spray-drying process is<br />

conceptually quite simple. Initially, the<br />

compound and a dispersion polymer are<br />

dissolved in a volatile organic solvent<br />

(e.g., acetone or methanol). The solution is<br />

then introduced into the drying chamber<br />

of a spray dryer along with heated<br />

F I G U R E 2<br />

nitrogen. The heated gas evaporates the<br />

solvent, leaving a dried powder, which is<br />

collected in a cyclone or baghouse.<br />

Of course, the process isn’t quite that<br />

simple. If the drying capacity of the<br />

nitrogen is insufficient, product will be<br />

deposited on the walls of the spray dryer,<br />

resulting in “spray-painting” and reduction<br />

of product yield. If the drying capacity of<br />

the nitrogen is overdesigned, energy costs<br />

are increased, and the risk of degrading a<br />

thermally labile compound is increased.<br />

However, by using best practices,<br />

spray-drying conditions can be identified<br />

that allow the product particles to be<br />

maintained at low temperatures (due to<br />

evaporative cooling as the volatile solvent<br />

evaporates), while producing high product<br />

yields (avoiding spray-painting of the<br />

walls of the dryer). These best practices<br />

for spray-drying process development<br />

were highlighted in a 2009 publication in<br />

the Journal of Pharmaceutical Innovation.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

27


COMPOUND PROPERTIES:<br />

AN IMPORTANT PART OF<br />

THE EQUATION<br />

Compounds with a wide variety of<br />

physical-chemical properties can be<br />

formulated as amorphous dispersions.<br />

During our decade-and-a-half formulating<br />

amorphous dispersions, we have found<br />

fewer than 10 compounds that could not<br />

be formulated as amorphous dispersions.<br />

These fall into two categories: (1) highly<br />

reactive compounds that chemically<br />

degrade in the amorphous state and<br />

(2) compounds with very high melting<br />

points that have poor physical stability or<br />

poor solubility in suitable spray solvents.<br />

However, this small group of compounds<br />

appears to be the exception rather than the<br />

rule.<br />

The vast majority of compounds we<br />

have worked on (out of more than 500<br />

tested) are amenable to formulation as<br />

amorphous dispersions, although their<br />

physical-chemical properties span meltingpoint<br />

ranges from 0°C to more than 350°C<br />

and log P values from less than 0 to more<br />

than 10. However, formulation<br />

compromises must be made for<br />

compounds at the extremes of physicalchemical<br />

property ranges to ensure<br />

performance is maintained. Generally,<br />

these compromises involve decreasing the<br />

amorphous dispersion drug loading or<br />

“swamping out” the poor drug properties<br />

by diluting the drug with the dispersion<br />

polymer. Of course, diluting the drug in<br />

polymer makes obtaining higher drug<br />

doses increasingly difficult. This is<br />

especially challenging in therapeutic areas<br />

such as anti-infectives, antivirals, and<br />

28<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

oncology, in which high doses are<br />

typically needed. To address these classes<br />

of compounds, we have developed<br />

alternative technologies.<br />

These alternatives include a<br />

nanoadsorbate technology, in which a<br />

dispersion is coated on a high surface area<br />

support to increase the dissolution rate.<br />

This technology has been used in multiple<br />

clinical trials and notably has been used in<br />

the clinic to give dose-linear absorption of<br />

a log P 10 compound with a solubility of<br />

less than 10 ng/mL. To address the other<br />

extreme–compounds with very high<br />

melting points and a strong tendency to<br />

crystallize in both the solid state and<br />

solution–we have developed a crystallized<br />

dispersion technology in which small,<br />

tens-of-nanometer-sized crystals are<br />

intentionally formed in the dispersion.<br />

This approach maintains a high-energy<br />

form of the compound and prevents<br />

further crystallization of the compound to<br />

a lower-energy species. This technology<br />

has also been used to advance a number of<br />

compounds to clinical evaluation.<br />

THE IMPORTANCE OF<br />

EXCIPIENTS<br />

Excipient choice is critical and<br />

depends on the specific solubilization<br />

problem being solved. Recently, BCS<br />

Class II compounds have been<br />

subcategorized into DCS IIA and DCS IIB<br />

classes by Professor Dressman and coworkers<br />

from GlaxoSmithKline. DCS<br />

Class IIA compounds have inadequate<br />

dissolution rates, whereas DCS Class IIB<br />

compounds are truly solubility limited,<br />

making bioavailability difficult to achieve<br />

without concentration enhancing<br />

polymers. An added complication is that<br />

some compounds formulated as<br />

amorphous dispersions dissolve into<br />

solution and stay at that amorphous<br />

concentration. Others, particularly those<br />

with high melting points, dissolve and<br />

then rapidly crystallize if they are not<br />

sustained by the polymer.<br />

As an example, for a DCS Class IIA<br />

compound for which amorphous solubility<br />

is easily sustained, the critical problem to<br />

solve is getting the compound to rapidly<br />

dissolve. Often, neutral polymers, such as<br />

BASF’s Kollidon VA64 (vinyl pyrrolidonevinyl<br />

acetate copolymers), work well due<br />

to their high water solubility, ability to<br />

dissolve in gastric media, and the reduced<br />

need to sustain the drug concentration. In<br />

fact, Kollidon VA64 can be highly<br />

preferred in these cases due to its high<br />

solubility in organic solvents typically<br />

used in spray-drying. Higher spray-solvent<br />

concentrations can substantially improve<br />

throughputs, which can lead to a decrease<br />

in the cost of goods.<br />

For compounds that have a tendency<br />

to crystallize in solution or the solid state<br />

and that require improved solubilization,<br />

enteric, cellulosic polymers, such as Dow<br />

or Shin Etsu’s hydroxypropyl<br />

methylcellulose acetate succinate<br />

(HPMCAS) or Eastman’s cellulose acetate<br />

phthalate (CAP), perform very well. This<br />

is due to a combination of factors–one<br />

being that the high glass-transition<br />

temperature of the polymers lead to superb<br />

solid-state stability for the dispersions; the<br />

second is that both polymers form colloids<br />

in solution that combine with the drug to


yield both enhanced solubility and rapid<br />

dissolution rates.<br />

As with technology, we are not tied to<br />

the choice of specific excipients, but we<br />

do find that HPMCAS has led to superior<br />

performance for more than half of the<br />

compounds we have worked on. An added<br />

benefit of formulating with HPMCAS is<br />

that Bend Research and its clients have<br />

access to one of the most complete safety<br />

packages through the Type V drug master<br />

file (DMF) for this polymer.<br />

THE FUTURE OF<br />

SOLUBILIZATION<br />

TECHNOLOGY<br />

With BCS Class II compounds<br />

comprising an increasing percentage of<br />

compounds in our clients' discovery<br />

portfolios, Bend Research is positioned to<br />

continue to provide the highest-quality<br />

support and innovation for our clients'<br />

pipelines. This proactive support is<br />

focused on two crucial areas: (1) support<br />

of the existing technology platforms and<br />

(2) development of next-generation<br />

technologies.<br />

To support existing technologies, we<br />

are engaged in ensuring clients have<br />

seamless access to the spray-drying<br />

capacity required as their compounds<br />

advance, as well as ensuring high-quality<br />

excipient supplies. An example of ready<br />

client access to spray-drying capacity is<br />

reflected in our collaboration with<br />

Hovione. This collaboration allows for<br />

facile transfer of programs from Bend<br />

Research’s research and development<br />

facilities to Hovione commercial facilities,<br />

either during Phase III clinical trials or<br />

following product launch. We know from<br />

experience that each client has preferred<br />

timing for this transfer, and we work with<br />

them to identify the best options and to<br />

perform technology transfer against this<br />

plan.<br />

Similarly, we are working with the<br />

leading excipient providers to ensure<br />

access to a reliable supply of<br />

solubilization excipients that meet the<br />

critical-to-quality properties that are keys<br />

to their performance. These relationships<br />

are an active piece of our technologydevelopment<br />

portfolio and continue to<br />

mature.<br />

In addition to supporting the existing<br />

technologies, Bend Research is working to<br />

develop new solubilization technologies.<br />

These efforts include working with the<br />

excipient providers to develop new<br />

solubilization excipients that improve<br />

performance and processability as well as<br />

developing improved spray-drying<br />

processes and equipment.<br />

Finally, in addition to our internal<br />

research and development efforts and our<br />

ongoing work with our alliance partners,<br />

we are actively forming partnerships to<br />

commercialize the best new solubilization<br />

technologies in partnership with<br />

universities and small companies. Based<br />

on our company’s successful track record<br />

in advancing numerous technologies and<br />

reducing them to practice, this work is<br />

only natural, particularly given the<br />

increasing number of poorly soluble<br />

compounds being developed by the<br />

pharmaceutical industry. u<br />

Pr<br />

B I O G R A P H Y<br />

Dr. Rod Ray is Chief Executive Officer and<br />

Chairman of the Board at Bend Research,<br />

where he has worked since 1983. During his<br />

time at Bend Research, Dr. Ray has held<br />

numerous positions specializing in the<br />

development and commercialization of a wide<br />

range of products. He has been instrumental<br />

in directing the management of large-scale<br />

programs to advance pharmaceutical<br />

compounds through the development process<br />

to commercialization, serving as the primary<br />

management contact for client companies. In<br />

addition to his expertise in advancing<br />

pharmaceutical processes and products, Dr.<br />

Ray has extensive experience in<br />

commercializing diverse products for the<br />

electronics, energy, medical, agricultural, and<br />

space industries. He earned his BS in Chemical<br />

Engineering from Oregon State University, and<br />

his MS and PhD in Chemical Engineering from<br />

the University of Colorado - Boulder. He is a<br />

licensed Professional Engineer in Colorado and<br />

Oregon. Dr. Ray holds 21 US patents and has<br />

41 scientific publications to his credit.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

29


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

30<br />

TRANSDERMAL<br />

D E L I V E R Y<br />

Recent <strong>Development</strong>s in Microneedle<br />

Technology for Transdermal <strong>Drug</strong><br />

<strong>Delivery</strong> & Vaccination<br />

By: Mikolaj Milewski and Amitava Mitra<br />

MICRONEEDLE-ASSISTED<br />

TRANSDERMAL DRUG<br />

DELIVERY<br />

Dissolving Microneedles<br />

These types of microneedles are<br />

fabricated using biodegradable polymers in<br />

which drugs or vaccines are encapsulated<br />

in the microneedles. 5 Once in the skin, the<br />

microneedles dissolve, thus releasing the<br />

drug. The enhancement of flux afforded by<br />

microneedles has generated significant<br />

interest in this transdermal delivery<br />

technology for delivery of peptides and<br />

proteins. Fukushima et al reported using<br />

two-layered dissolving microneedles for<br />

transdermal delivery of human growth<br />

hormone (rhGH) and desmopressin<br />

<strong>IN</strong>TRODUCTION<br />

The skin has long been recognized as a potential target for local and systemic drug delivery as well as vaccination. The<br />

practical value of transdermal route of drug administration is, however, limited by substantial barrier properties of the skin.<br />

Physiologically beneficial protection that the skin provides against xenobiotic permeation is a drawback from the perspective<br />

of percutaneous transport of therapeutic agents. Thus, a delivery system that temporarily and reversibly permeabilizes the<br />

skin can enable delivery of a wide spectrum of molecules across the skin. One such technology is microneedles, which are<br />

micron-scale needles that can create microscopic pores in the stratum corneum and upper layers of the epidermis, thereby<br />

enhancing skin permeation up to several orders of magnitude. 1 Four designs of microneedles have been developed: dissolving<br />

microneedles made of biodegradable polymers that encapsulate drugs or vaccines, solid coated microneedles in which the<br />

drugs or vaccines are coated on the microneedle surface, hollow microneedles for injection, and solid microneedles to pierce<br />

the skin followed by application of a drug patch (poke and patch approach) (Figure 1). 2 The purpose of this article is to<br />

highlight some of the recent advances in the field of microneedle-mediated transdermal drug delivery and vaccination,<br />

including summaries of preclinical pharmacokinetic data and a brief overview of clinical studies. This review encompasses the<br />

time period from 2010 to present day. For more background on microneedles, interested readers should refer to the following<br />

references. 3,4<br />

(DDAVP) in rats. 6 rhGH (22 kDa) was<br />

formulated at 2-microgram doses in the<br />

dissolving microneedles composed of<br />

sodium chondroitin sulfate and dextran.<br />

The application of this microneedle<br />

formulation to the rat abdomen produced a<br />

PK profile characterized by fast attainment<br />

of peak concentration (t max = 15 mins) and<br />

gradual decrease in the plasma rhGH level<br />

with terminal half-life approximating 25<br />

mins. Chondroitin-based and dextran-<br />

based microneedles performed similarly.<br />

Importantly, the authors observed dose-<br />

proportional increase in C max , and the area<br />

under concentration-time curve (AUC) as<br />

a function of dose. Also, the bioavailability<br />

was very high and amounted to<br />

approximately 95% in chondroinin<br />

microneedles and 73% in dextran<br />

microneedles. Interestingly, the IV bolus<br />

injection of rhGH revealed much shorter<br />

elimination half-life (4 mins) implying<br />

flip-flop kinetics for microneedle-mediated<br />

delivery. Hence, the terminal half-life of<br />

25 mins following microneedle application<br />

was attributed to the absorption rather than<br />

elimination phase. On the other hand,<br />

DDAVP (1.07 kDa) chondroitin<br />

microneedles showed no flip-flop kinetics<br />

and an absorption phase half-life of 14<br />

mins. PK profiles were characterized by<br />

tmax of 30 mins and terminal half-life of<br />

approximately 2 hrs. Approximately 0.1<br />

mg rhGH could be formulated into a patch<br />

of 100 microneedles. The microneedle<br />

formulations were stable for at least 1


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

32<br />

month under refrigeration or freezing<br />

conditions.<br />

F I G U R E 1<br />

A schematic depiction of four basic modes of<br />

microneedle application in transdermal drug<br />

delivery: a) poke and patch in which the skin<br />

is pretreated with microneedles to be<br />

subsequently covered by a transdermal<br />

formulation; b) coated microneedles in which<br />

drug is coated on the surface of the<br />

microneedles; c) dissolving microneedles in<br />

which drug is embedded in the matrix of<br />

dissolving microneedles (typically polymer- or<br />

sugar-based); and d) hollow microneedles in<br />

which drug formulation is injected<br />

intradermally. Reproduced with permission<br />

from the International Journal of<br />

Pharmaceutics. 2008;364:227-236.<br />

An independent, but related, study of hGH<br />

in rats was carried out by Lee et al. 7<br />

Carboxymethylcellulose-(CMC) and CMC-<br />

trehalose-based dissolving microneedles were<br />

administered to hairless rats reaching C max after<br />

approximately 30 mins and subsequently,<br />

plasma concentration decreased gradually with<br />

a half-life of 1.1 hrs. The authors subsequently<br />

compared microneedle-mediated hGH delivery<br />

with subcutaneous (SC) injections<br />

demonstrating markedly similar PK profiles.<br />

Another study by Yukako et al reported<br />

low bioavailability of peptide leuprolide acetate<br />

(LA, 1.2 kDa) following administration to rats<br />

by dissolving microneedles and subcutaneous<br />

injection. 8 In accordance with in vitro release<br />

data, in vivo PK profiles demonstrated short<br />

tmax (15 mins) for microneedles and 20 mins<br />

for SC injection. However, low bioavailabilities<br />

(32%) were observed due to the metabolic<br />

instability of LA in skin. This study highlights<br />

the potential limitations of microneedle-<br />

mediated drug delivery related to the<br />

physiology of the skin rather than the<br />

microneedle technology itself.<br />

Coated Microneedles<br />

Daddona et al studied the<br />

pharmacokinetics and pharmacodynamics of<br />

parathyroid hormone (PTH, 4.1 kDa)<br />

microneedle-mediated delivery in humans. 9<br />

The microneedle arrays consisted of titanium<br />

microneedles coated with PTH (20 to 40<br />

micrograms) attached to an adhesive patch. The<br />

patch is applied with a hand-held and reusable<br />

applicator. A once-daily subcutaneous injection<br />

of FORTEO ® , which is used as a therapy for<br />

advanced osteoporosis in men and post-<br />

menopausal women, served as a reference for<br />

the evaluation of the coated microneedle<br />

system performance with a wear time of 30<br />

mins. Clinical studies in post-menopausal<br />

women demonstrated that the microneedle<br />

system achieved shorter t max of approximately 8<br />

mins compared to 24 mins for SC injection.<br />

The terminal half-life after SC injection was<br />

longer compared to microneedle delivery, and<br />

implied flip-flop kinetics. The relative<br />

bioavailability of the PTH microneedle patch<br />

ranged from 40% to 80%. Interestingly, these<br />

differences were believed to be dictated by<br />

application to different anatomical body sites<br />

and not by the patch performance<br />

inconsistency, with the highest relative<br />

exposure that was obtained from the abdomen,<br />

followed by upper arm, and the lowest from the<br />

thigh. In each case, the residual PTH found on<br />

the microneedle array after application was<br />

< 20%. Dose-proportional increase in the AUC<br />

was observed in the clinic. The inter-subject<br />

and between-occasion intra-subject variability<br />

seen in the PTH-patch and FORTEO were<br />

comparable. Pharmacodynamically, the PTH-<br />

coated microneedle produced dose-proportional<br />

increase in the bone mineral density. The<br />

magnitude of this effect was higher compared<br />

to FORTEO and might be related to different<br />

PK achieved with the application of PTH<br />

microneedle. Moisture and oxygen-devoid<br />

packaged PTH microneedle patches were<br />

found to be stable in room temperature for 2<br />

years, which is a significant advantage over<br />

FORTEO that needs to be stored at 5°C to 8°C.<br />

Another study involving the Zosano<br />

microneedle patch system was carried out by<br />

Peters et al. 10 In this study, the stability and<br />

preclinical performance of erythropoietin-<br />

coated microneedles (EPO, approx. 34 kDa)<br />

was evaluated in rats. Pharmacokinetic profiles<br />

obtained following SC and microneedle-<br />

assisted administration of EPO were alike and<br />

resulted in tmax of 6 to 12 hrs and a terminal<br />

elimination half-life of 9 to 12 hrs. A linear<br />

AUC dose-response curve was achieved within<br />

the 7- to 200-microgram dose range tested.<br />

Moreover, the relative bioavailability of EPO<br />

after microneedle administration was<br />

comparable to that obtained following SC<br />

injection.<br />

Zhang et al investigated the potential of<br />

lidocaine-coated microneedles for local<br />

analgesic action in domestic swine. 11 This<br />

study was unique because here, an attempt was<br />

made to use the microneedles to enhance local<br />

(dermal) delivery of a therapeutic agent. The<br />

authors used 3M’s 500 micrometer-long solid<br />

microneedles, termed sMTS, dip-coated with<br />

aqueous lidocaine (234 Da) solution. The local<br />

lidocaine concentration, obtained at the<br />

treatment site immediately following<br />

microneedle application was higher than the<br />

estimated level needed for analgesia and was<br />

maintained for an hour when co-administered<br />

with vasoconstrictive lidocaine.<br />

Hollow Microneedles<br />

Harvey et al investigated microneedlebased<br />

intradermal and subcutaneous delivery of<br />

protein drugs in Yucatan minipig. 12 The authors<br />

employed a single microneedle device for<br />

injection of insulin and somatropin and a threemicroneedle<br />

device to inject etanercept in the<br />

dermal space. In the context of this study,<br />

microneedle refers to a manually assembled<br />

complex of 1-mm-long 34-gauge steel needle,<br />

flexible tubing, and an analytical microsyringe<br />

drug reservoir. Formulation volumes injected<br />

intradermally varied in the range of 50 to 250<br />

microliters. Etanercept (132 kDa) injections<br />

demonstrated markedly shortened tmax (5 hrs) of


microneedle-mediated delivery as compared to<br />

18 hrs for subcutaneous injection. The absolute<br />

bioavailability was 75% for microneedles and<br />

50% for SC administration. Similarly,<br />

somatropin (22 kDa) injections showed faster<br />

absorption kinetics following intradermal<br />

microneedle-mediated injections (t max = 30<br />

mins) as compared to subcutaneous<br />

administration (t max = 2.75 hrs). The absolute<br />

bioavailability was found to be 100% for both<br />

routes. Moreover, insulin lispro (5.8 kDa)<br />

demonstrated rapid uptake to systemic<br />

circulation when administered intradermally<br />

(t max = 22 mins) and slower uptake following<br />

SC injection (t max = 61 mins). Interestingly, the<br />

absorption rate of regular and fast-acting<br />

insulin after microneedle-mediated intradermal<br />

injection was comparable. In all of the<br />

aforementioned studies, shorter t max was<br />

accompanied by higher C max . The authors<br />

performed additional imaging studies that led<br />

to the hypothesis that rapid uptake seen for<br />

microneedle-mediated intradermal protein<br />

delivery is aided by fast lymphatic uptake in<br />

the dermis.<br />

Pettis et al studied intradermal<br />

microneedle delivery of insulin lispro (5.8<br />

kDa) versus its SC delivery in healthy human<br />

volunteers. 13 Application of insulin<br />

intradermally through a single stainless steel<br />

hollow microneedle and an SC injection<br />

resulted in rapid systemic absorption and<br />

bioavailability of microneedle-mediated<br />

delivery comparable to SC injection. T max<br />

values increased (36 mins, 40 mins, and 46<br />

mins) with increasing microneedle length (1.25<br />

to 1.5 to 1.75 mm) and proved to be the highest<br />

for SC injection (64 mins).<br />

Subcutaneous and intradermal delivery of<br />

liquid formulations through multiple hollow<br />

microneedles (hMTS, 3M <strong>Drug</strong> <strong>Delivery</strong><br />

Systems) in domestic swine was studied by<br />

Burton et al. 14 Pharmacokinetic studies<br />

compared microneedle-mediated and SC<br />

administration of three model compounds:<br />

naloxone (322 Da) hydrochloride, hGH<br />

(22kDa), and equine tetanus anti-toxin (ETAT,<br />

approx. 150 kDa). Interestingly, naloxone<br />

hydrochloride showed faster absorption<br />

following SC injection compared to<br />

microneedle administration. In contrast, hGH<br />

showed faster absorption following<br />

microneedle administration. The antibody<br />

(ETAT) showed similar PK profiles with both<br />

SC and microneedle administration.<br />

Poke & Patch<br />

This delivery approach involves piercing<br />

the upper layers of the skin with solid<br />

microneedles followed by application of a drug<br />

formulation (eg, patch, gel) at that site. 2 The<br />

pretreatment creates microscopic pores in the<br />

skin, thereby enhancing flux of the molecules.<br />

Zhang et al reported increased bioavailability<br />

of L-carnitine (LC) in rats with the poke and<br />

patch method compared to its oral<br />

administration. 15 Although LC is a small<br />

molecular weight compound (161 Da), it does<br />

not permeate at high rates through intestinal<br />

epithelium or skin due to its ionized and<br />

hydrophilic nature. Authors investigated in<br />

vitro permeation of LC from solution across<br />

untreated and microneedle pretreated skin and<br />

demonstrated a 17-fold increase in flux across<br />

skin pretreated with microneedles.<br />

Subsequently, several carbomer hydrogels were<br />

evaluated as formulation options. The CP980<br />

gel showed an LC transport rate of 72% of that<br />

of the aqueous solution formulation across<br />

microneedle pretreated skin. Finally, a rat PK<br />

study comparing IV, oral, and poke and patch<br />

delivery methods was conducted. Oral delivery<br />

of LC resulted in only 8% bioavailability and<br />

t max of 2 hrs. Following a 6-hr microneedle-<br />

enhanced transdermal LC delivery, 22%<br />

bioavailability was achieved with t max at 4 hrs.<br />

The bioavailability was calculated on the basis<br />

of the total dose applied in the patch. An<br />

alternative calculation based on the fraction of<br />

the total dose that was actually delivered into<br />

skin yields a bioavailability of 81%.<br />

Interestingly, the poke and patch PK profile<br />

resembled a traditional non-microneedle<br />

percutaneous profile in the sense that relatively<br />

steady plasma levels were obtained in the 2- to<br />

6-hr time window. However, the likely reason<br />

for lack of more pronounced C max is that the<br />

timeline of the experiment was relatively short,<br />

and the microchannels did not close enough to<br />

significantly limit in vivo flux through the<br />

microchannels.<br />

F I G U R E 2<br />

Becton Dickinson's Micro Injection System<br />

demonstrating: a) microneedle shield; b)<br />

microneedle pre-attached to the tip of the<br />

syringe; c) vaccine-level check window; d)<br />

finger pads; and (e) plunger rod.<br />

Reproduced with permission from Vaccine 25<br />

(2007) 8833–8842.<br />

MICRONEEDLE-ASSISTED<br />

VACC<strong>IN</strong>ATION<br />

The use of the skin as a target site for<br />

vaccination has been largely limited due to the<br />

difficulty in reliably performing intradermal<br />

injections. With the advent of microneedles, a<br />

minimally invasive and precise intradermal<br />

placement of vaccine has become possible. The<br />

skin is known to be a highly immunogenic<br />

tissue containing a wealth of antigen-presenting<br />

cells, including epidermal Langerhans' cells<br />

and dermal dendritic cells. 16 As such, it is a<br />

good vaccination target with additional dosesparing<br />

potential compared to lessimmunogenic<br />

muscle tissue.<br />

A recent study by Weldon et al examined<br />

an influenza vaccine-coated microneedle in<br />

mice. 17 The authors chose to utilize trehalosestabilized,<br />

solubilized viral protein antigens<br />

rather than more widely employed inactivated<br />

influenza virus and virus-like particles.<br />

Microneedles coated with stabilized<br />

recombinant trimeric soluble hemagglutinin<br />

33<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

(sHA) provided superior protection against<br />

influenza virus compared to the unmodified<br />

sHA. Moreover, post-challenge lung titers<br />

demonstrated that intradermal vaccination<br />

resulted in greater clearance of replicating<br />

virus compared to the SC vaccination.<br />

In another study, Kommareddy et al<br />

investigated the use of dissolvable microneedle<br />

patches delivering influenza vaccine antigens in<br />

mice. 18 The authors employed TheraJect TM<br />

VaxMat TM microneedle technology to<br />

incorporate microgram quantities of vaccine in<br />

a microneedle patch. A set of in vitro and in<br />

vivo studies confirmed the integrity and<br />

immunogenicity of the antigens incorporated<br />

into a dissolving microneedle patch.<br />

However, perhaps most interestingly, 2011<br />

proved a ground-breaking year for the<br />

commercialization of microneedle drug delivery<br />

technologies in the US. On May 9, 2011, the<br />

FDA approved a first, and so far the only,<br />

microneedle-based product: Fluzone<br />

Intradermal ® influenza virus vaccine (Sanofi<br />

Pasteur). 19 This constituted a major milestone in<br />

the transition of microneedle systems from the<br />

developmental stage to the market place. The<br />

vaccine is administered from a prefilled<br />

microinjection system consisting of a custom<br />

syringe with a 30-gauge, 1.5-mm-long, hollow<br />

single microneedle developed by Beckton<br />

Dickinson (Figure 2). 20 The microneedle is about<br />

10 times shorter than a traditional needle used to<br />

perform intramuscular injections of Fluzone.<br />

Moreover, Phase III clinical data confirmed that<br />

intradermal delivery of vaccine allows for dose-<br />

sparing. A 9-microgram dose of vaccine<br />

delivered intradermally proved to be<br />

immunogenetically comparable to a 15-<br />

microgram dose delivered intramuscularly. 21<br />

Hence, a 40% reduction in the dose was<br />

achieved. In 2009, Intanza ® , an influenza vaccine<br />

based on the same microneedle technology, was<br />

approved by the European Medicines Agency<br />

(EMA). 22 These recent developments and the<br />

appearance of first products on the market have<br />

encouraged academic institutions and<br />

pharmaceutical industries to pursue the<br />

microneedle-based strategy for drug delivery and<br />

vaccination purposes as reflected by several<br />

34<br />

ongoing clinical trials.<br />

CL<strong>IN</strong>ICAL TRIALS <strong>IN</strong>VOLV<strong>IN</strong>G<br />

MICRONEEDLES<br />

A number of ongoing trails with<br />

microneedle-based delivery systems illustrate<br />

the significant interest in this new transdermal<br />

delivery technology. Several clinical studies are<br />

listed in ClinicalTrials.gov. 23 A few of the<br />

notable studies are discussed here. Sanofi<br />

Pasteur has completed a Phase II trial that<br />

assessed non-inferiority of fractional doses of<br />

IMOVAX ® Polio administered intradermally<br />

versus full doses of IMOVAX Polio<br />

administered intramuscularly (study results<br />

pending). Phase II and III studies sponsored by<br />

Emory University currently investigate the<br />

difference in glycemic control between the SC<br />

insulin catheter and microneedle for bolus<br />

delivery of insulin. NanoPass technologies'<br />

ongoing studies evaluate the safety and<br />

efficacy of a MicronJet microneedle device as<br />

well as safety, pharmacokinetics, and<br />

pharmacodynamics of insulin injected via<br />

MicronJet. A Phase II study on<br />

pharmacokinetics and pharmacodynamics of<br />

basal insulin infusion administered either<br />

intradermally or subcutaneously has been<br />

completed by Becton Dickinson (study results<br />

pending). Furthermore, a Phase I clinical study<br />

assessing tolerability of the application of a 3M<br />

microstructure transdermal system has been<br />

completed (study results pending). Finally,<br />

Zosano Pharma microneedle-based PTH patch<br />

had completed Phase II clinical trials, and the<br />

results were published and summarized in the<br />

aforementioned Coated Microneedles section.<br />

SUMMARY<br />

Early research in the area of microneedle-<br />

assisted transdermal drug delivery provided<br />

ample proof of its potential in enhancing and<br />

enabling transport of pharmaceuticals across<br />

thr skin. It also demonstrated its limitations<br />

related to the relatively small drug doses that<br />

can be used in conjunction with the<br />

microneedle technique and the microneedle<br />

fabrication complexity, including related<br />

challenges in therapeutic agents' stability.<br />

Following extensive investigation of<br />

microneedle fabrication methods, later studies<br />

focused toward improving the performance of<br />

existing microneedle systems. Multiple<br />

preclinical and clinical studies demonstrated<br />

their successful application in vivo. The<br />

viability of the microchannels was assessed in<br />

vivo and its implications for the applicability of<br />

the poke and patch method were recognized.<br />

Clinical successes have resulted in significant<br />

interest from both academia and the<br />

pharmaceutical industry in this delivery area. A<br />

selective review of the recent advancements in<br />

this field reported herein highlights growing<br />

sophistication of this technology in its ability to<br />

deliver a wide variety of molecules and<br />

vaccines with precision. Importantly, at a time<br />

when biopharmaceuticals (biologics and<br />

vaccines) are becoming an integral part of<br />

pharmaceutical pipelines, microneedle-based<br />

devices offer a promising alternative to<br />

traditional SC and intramuscular injections.u<br />

REFERENCES<br />

1. Henry S, McAllister DV, Allen MG,<br />

Prausnitz MR. Microfabricated<br />

microneedles: a novel approach to<br />

transdermal drug delivery. J Pharmaceut<br />

Sci. 1998;87(8):922-925.<br />

2. Arora A, Prausnitz MR, Mitragotri S.<br />

Micro-scale devices for transdermal drug<br />

delivery. Int J Pharm. 2008;364(2):227-236.<br />

3. Sachdeva V, Banga A. Microneedles and<br />

their applications. Recent Patents on <strong>Drug</strong><br />

<strong>Delivery</strong> & Formulation. 2011;5(2):95-132.<br />

4. Donnelly RF, Raj Singh TR, Woolfson AD.<br />

Microneedle-based drug delivery systems:<br />

microfabrication, drug delivery, and safety.<br />

<strong>Drug</strong> Deliv. 2010;17(4):187-207.<br />

5. Park J-H, Allen MG, Prausnitz MR.<br />

Biodegradable polymer microneedles:<br />

fabrication, mechanics, and transdermal<br />

drug delivery. J Controlled Rel.<br />

2005;104(1):51-66.<br />

6. Fukushima K, Ise A, Morita H, Hasegawa<br />

R, Ito Y, Sugioka N, Takada K. Two-layered<br />

dissolving microneedles for percutaneous<br />

delivery of peptide/protein drugs in rats.


Pharm Res. 2011;28(1):7-21.<br />

7. Lee JW, Choi SO, Felner EI, Prausnitz MR.<br />

Dissolving microneedle patch for<br />

transdermal delivery of human growth<br />

hormone. Small. 2011;7(4):531-539.<br />

8. Ito Y, Murano H, Hamasaki N, Fukushima<br />

K, Takada K. Incidence of low<br />

bioavailability of leuprolide acetate after<br />

percutaneous administration to rats by<br />

dissolving microneedles. Int J Pharm.<br />

2011;407(1-2):126-131.<br />

9. Daddona PE, Matriano JA, Mandema J,<br />

Maa YF. Parathyroid hormone (1-34)-coated<br />

microneedle patch system: clinical<br />

pharmacokinetics and pharmacodynamics<br />

for treatment of osteoporosis. Pharm Res.<br />

2011;28(1):159-165.<br />

10. Peters EE, Ameri M, Wang X, Maa YF,<br />

Daddona PE. Erythropoietin-coated ZP-<br />

microneedle transdermal system:<br />

preclinical formulation, stability, and<br />

delivery. Pharm Res. 2012 [Epub ahead of<br />

print].<br />

11. Zhang Y, Brown K, Siebenaler K,<br />

Determan A, Dohmeier D, Hansen K.<br />

<strong>Development</strong> of lidocaine-coated<br />

microneedle product for rapid, safe, and<br />

prolonged local analgesic action. Pharm<br />

Res. 2011;29(1):170-177.<br />

12. Harvey J, Kaestner SA, Sutter DE, Harvey<br />

NG, Mikszta JA, Pettis RJ. Microneedle-<br />

based intradermal delivery enables rapid<br />

lymphatic uptake and distribution of<br />

protein drugs. Pharmaceeut Res.<br />

2011;28(1):107-116.<br />

13. Pettis J, Ginsberg B, Hirsch L, Sutter D,<br />

Keith S, McVey E, Harvey Noel G,<br />

Hompesch M, Nosek L, Kapitza C,<br />

Heinemann L. Intradermal microneedle<br />

delivery of insulin lispro achieves faster<br />

insulin absorption and insulin action than<br />

subcutaneous injection. Diabetes Technol<br />

Therapeut. 2011;13(4):435-442.<br />

14. Burton A, Ng C-Y, Simmers R, Moeckly<br />

C, Brandwein D, Gilbert T, Johnson N,<br />

Brown K, Alston T, Prochnow G,<br />

Siebenaler K, Hansen K. Rapid<br />

intradermal delivery of liquid formulations<br />

using a hollow microstructured array.<br />

Pharmaceut Res. 2010;28(1):31-40.<br />

15. Zhang S, Qin G, Wu Y, Gao Y, Qiu Y, Li F,<br />

Xu B. Enhanced bioavailability of Lcarnitine<br />

after painless intradermal<br />

delivery vs. oral administration in rats.<br />

Pharm Res. 2011;28(1):117-123.<br />

16. Kupper TS, Fuhlbrigge RC. Immune<br />

surveillance in the skin: mechanisms and<br />

clinical consequences. Nat Rev Immunol.<br />

2004;4(3):211-222.<br />

17. Weldon WC, Martin MP, Zarnitsyn V,<br />

Wang B, Koutsonanos D, Skountzou I,<br />

Prausnitz MR, Compans RW. Microneedle<br />

vaccination with stabilized recombinant<br />

influenza virus hemagglutinin induces<br />

improved protective immunity. Clin<br />

Vaccine Immunol. 2012;18(4):647-654.<br />

18. Kommareddy S, Baudner BC, Oh S, Kwon<br />

SY, Singh M, O'Hagan DT. Dissolvable<br />

microneedle patches for the delivery of<br />

cell-culture-derived influenza vaccine<br />

antigens. J Pharm Sci. 2012;101(3):1021-<br />

1027.<br />

19. 2009. FDA Approval Letter - Fluzone<br />

Intradermal;<br />

http://www.fda.gov/BiologicsBloodVaccine<br />

s/Vaccines/ApprovedProducts/ucm255160.<br />

htm. ed. FDA.<br />

20. Laurent PE, Bonnet S, Alchas P, Regolini<br />

P, Mikszta JA, Pettis R, Harvey NG.<br />

Evaluation of the clinical performance of a<br />

new intradermal vaccine administration<br />

technique and associated delivery system.<br />

Vaccine. 2007;25(52):8833-8842.<br />

21. 2011. Fluzone Intradermal(R) prescribing<br />

information;<br />

https://www.vaccineshoppe.com/image.cf<br />

m?doc_id=12427&image_type=product_p<br />

df. ed.: Sanofi Pasteur.<br />

22. 2009 Intanza market authorisation;<br />

http://www.ema.europa.eu/ema/index.jsp?c<br />

url=pages/medicines/human/medicines/000<br />

957/human_med_000842.jsp&mid=WC0b<br />

01ac058001d124&murl=menus/medicines/<br />

medicines.jsp&jsenabled=true. ed.: EMA.<br />

23. ClinicalTrials.gov; http://clinicaltrials.gov/.<br />

ed.: U.S. National Library of Medicine.<br />

Pr<br />

B I O G R A P H I E S<br />

Dr. Mikolaj<br />

Milewski is a<br />

Senior Research<br />

Pharmacist at<br />

Merck. As a<br />

member of the<br />

Biopharmaceutics<br />

group, he<br />

assesses<br />

bioperformance<br />

risk of toxicology<br />

and clinical formulations. He also evaluates<br />

feasibility of alternative drug delivery routes<br />

for new chemical entities and existing<br />

products. He has authored research and review<br />

articles in peer-reviewed journals and is a<br />

member of AAPS and ACS. His areas of interest<br />

focus on oral and transdermal formulations,<br />

pharmacokinetics, and development of<br />

microneedle-based drug delivery systems. Prior<br />

to joining Merck, Dr. Milewski earned his PhD<br />

in Pharmaceutical Sciences from the University<br />

of Kentucky in Lexington.<br />

Dr. Amitava<br />

Mitra earned his<br />

BS in Pharmacy<br />

from Birla<br />

Institute of<br />

Technology, India,<br />

his PhD in<br />

Pharmaceutical<br />

Sciences from the<br />

University of<br />

Maryland, and<br />

completed his post-doctoral fellowship from<br />

Fox Chase Cancer Center. Dr. Mitra has<br />

published research articles in peer-reviewed<br />

journals and has authored review articles and<br />

book chapters. He has numerous podium and<br />

poster presentations in national and<br />

international conferences. He is a member of<br />

the AAPS, CRS, and the Rho Chi Pharmacy<br />

Honor Society. He is a recipient of the<br />

Controlled Release Society-3M <strong>Drug</strong> <strong>Delivery</strong><br />

Systems Graduate Student Outstanding<br />

Research Award in 2005 and American<br />

Association of Pharmaceutical Scientists-<br />

National Biotechnology Conference Graduate<br />

Student Award in 2006.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

35


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

36<br />

<strong>IN</strong>JECTION<br />

M O L D I N G<br />

Injection Molding in the<br />

Pharmaceutical Industry<br />

By: Andrew Loxley, PhD, and Brett Braker<br />

THE MOLDER<br />

The major components of an<br />

injection molder are shown in Figure 1.<br />

The IM process involves four essential<br />

steps:<br />

1. Melting of material<br />

2. Mass transfer of molten material<br />

from an injector into channels<br />

called “runners” in the mold and<br />

finally into the mold cavity<br />

3. Hardening of the material in the<br />

mold to the shape of the cavity<br />

4. Ejecting the part from the cavity<br />

to produce the final product<br />

The earliest injection molders used a<br />

simple piston to force molten material into<br />

the mold. Modern molders use a combined<br />

heated barrel/screw/ram assembly in which<br />

the solid material is fed to the hopper of<br />

the heated barrel, where it is melted by a<br />

<strong>IN</strong>TRODUCTION<br />

Many of the processes used to manufacture products within the pharmaceutical industry are unique to the particular<br />

product; however, there are also processes that have been borrowed and adapted from other manufacturing industries and<br />

successfully employed in the development of pharmaceutical products. One such example is injection molding (IM); a process<br />

developed in the late 19th century for the manufacture of simple plastic objects, such as combs, and later extended to all<br />

manner of parts made from thermoplastic and thermoset resins. Parts made by IM that are widely used in the pharmaceutical<br />

industry include caps, seals, closures, valves, syringes, inhalers, and the like. As with other plastic processing technologies<br />

that enable pharmaceutical solutions for otherwise difficult problems, IM is now gaining in popularity for manufacturing<br />

more complex device parts, and is even the platform of choice for preparing certain proprietary drug products.<br />

combination of the heat from the heater<br />

bands and the shear forces between the<br />

material, screw, and barrel. The molten<br />

material is conveyed by the screw toward<br />

the nozzle at the end of the barrel, and the<br />

screw then travels forward in the barrel as<br />

a ram to inject the material into the mold<br />

on each cycle. The addition of the screw<br />

also allows for some mixing so that<br />

multiple feedstocks can be added<br />

F I G U R E 1<br />

Diagram of basic injection molding machine.<br />

simultaneously to prepare for example<br />

colored parts, or to reuse scrap from<br />

previous runs. Two examples of pilot-scale<br />

injection molders from Nissei and Arburg<br />

are shown in Figure 2. AB Insturments,<br />

Thermo Haake and Alba are among<br />

makers of lab-scale injection molding<br />

units.<br />

Molders can be hydraulic, electric, or<br />

pneumatic, with electric or pneumatic


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

38<br />

being preferred for pharmaceutical applications<br />

due to the potential issues of clean room<br />

contamination from hydraulic fluid aerosols.<br />

IM machines range in sizes from tabletop<br />

versions making micro-parts a few millimeters<br />

in size or smaller, to large-scale production<br />

machines requiring clamp pressures of the<br />

order of thousands of tons to hold the mold<br />

halves together during the molding process.<br />

MATERIALS<br />

Materials that are solid at room<br />

temperature must be heated above the meting<br />

temperature (T m ) before being pumped into the<br />

mold cavity. The temperature of the<br />

thermoplastic material needs to be raised<br />

sufficiently above T m to reduce its melt<br />

viscosity and allow transfer from its reservoir<br />

to the mold cavity at manageable pressures.<br />

The higher the temperature above T m , the lower<br />

the melt viscosity and hence the lower the<br />

pressure required to pump the material through<br />

the runners into the cavity. Of course, elevated<br />

temperatures accelerate thermal degradation (of<br />

polymers, additives, and drugs), so the lowest<br />

temperature that allows for reproducible part<br />

production should be used. Thermoplastic<br />

materials with glass transition temperatures<br />

(T g ) above room temperature form hard parts<br />

upon cooling, and suitable materials include<br />

resins, such as polystyrene, poly(methyl<br />

methacrylate), polypropylene, and<br />

polyethylene. Thermoplastic materials with T g<br />

below room temperature (thermoplastic<br />

elastomers) form rubbery parts on cooling, and<br />

example materials suitable for IM are ethylene<br />

vinyl acetate copolymers (EVA, eg,<br />

VitalDose TM ), various polyurethanes (eg,<br />

Chronothane TM , Elasthane TM , Tecoflex ® ),<br />

polystyrene-polyisobutylene block terpolymers<br />

(eg, Kraton TM ), polyether-polyamide block<br />

copolymers (eg, Peebax ® ), and polyvinylbutyral<br />

(eg, Butvar ® ).<br />

When the material is a liquid at room<br />

temperature, and cures in the mold by a<br />

chemical reaction to form the final part, the<br />

process is called reactive IM (RIM) and is<br />

exemplified by silicone elastomers in which<br />

low molecular weight reactive silicone liquids<br />

are cured in the mold at elevated temperatures<br />

by Platinum-catalyzed or Tin-catalyzed<br />

crosslinking reactions. Cycle times are<br />

typically longer for silicones than for<br />

thermoplastic products, as the part must cure<br />

before being ejected from the mold, and this is<br />

usually slower than simple cooling.<br />

MOLDS<br />

Molds are made of metal plates that have<br />

precision machined cavities in which the part<br />

cools or cures to take its final form after the<br />

material is injected into it. At the smallest<br />

tabletop injection molder scale, molds are<br />

F I G U R E 2<br />

Examples of injection molding machines, on the left, a Nissei hydraulic unit, and on the right, an<br />

Arburg electric unit.<br />

F I G U R E 3<br />

mostly made from aluminum to save on costs<br />

and an example o-ring mold is shown in Figure<br />

3. Production molds for medical and<br />

pharmaceutical applications are made of<br />

stainless steel of appropriate regulatory grade.<br />

Depending on the size and complexity, the cost<br />

of molds can range from a few thousand<br />

dollars and can go up to several hundreds of<br />

thousands.<br />

The runners may be embedded within the<br />

plane of the cavity in the mold and filled with<br />

polymer from the injection nozzle on each<br />

cycle. Molten material is distributed to each<br />

cavity in the mold from the runners. Because<br />

the cavity is generally not heated, the design is<br />

A simple aluminum o-ring mold showing both mold halves. The cold-runner that feeds the cavity<br />

is the channel at the top of the upper-right half.


called a cold-runner mold, and material in the<br />

cold runner hardens or cures with the part and<br />

is then removed from the part and discarded as<br />

scrap or in the case of stable thermoplastic<br />

formulations, sent to be recycled in a later<br />

molding run after each cycle. Cold runner<br />

systems are generally used for applications in<br />

which materials are inexpensive or when use of<br />

recycled material is acceptable, as they are less<br />

expensive than hot runner systems. However,<br />

materials cannot be reprocessed indefinitely,<br />

especially if thermally labile, and if thermoset<br />

materials are used in cold runner molds, the<br />

material must be discarded.<br />

Hot runner systems use a heated manifold<br />

that is fed by the injection nozzle and keeps the<br />

material molten in the runners in the mold<br />

frame outside the plane of the cavity. The<br />

molten material enters the cavity from the<br />

runners via valve-gates or tips, and there is no<br />

scrap, so costly raw materials losses are<br />

minimized. Only the material in the cavity cools<br />

and hardens on each cycle, and molten material<br />

for the next cycle is forced into the cavity from<br />

the hot runners as fresh material is pumped into<br />

the hot manifold from the barrel nozzle. The<br />

fact that material remains continuously molten<br />

in the mold means that thermally sensitive<br />

materials can be subject to degradation, and the<br />

volume of hot runners should be kept as low as<br />

possible (a few cavity volumes at most) to<br />

F I G U R E 4<br />

In vitro release of dapivirine from an EVA matrix-type IVR made by injection molding (25 mg<br />

dapivirine).<br />

ensure labile material does not have a long<br />

residence time in the molten state.<br />

PHARMACEUTICAL PRODUCTS<br />

BY <strong>IN</strong>JECTION MOLD<strong>IN</strong>G<br />

Simple and complex shapes can be<br />

produced by IM, and as such, the process is<br />

used to prepare a wide variety of plastic<br />

medical device parts from caps, seals, closures,<br />

syringes, valves, and even implants. All of<br />

these require formulation of polymers with a<br />

range of additives, such as colorants,<br />

antioxidants, fillers, and plasticizers. Many of<br />

the compounds are pre-prepared by hot-melt<br />

extrusion, pelletized, and the pellets fed to the<br />

injection molder to form the part.<br />

Whereas the halves of a gelatin capsule<br />

that can be filled with API formulation are<br />

traditionally made by hardening a gelatin<br />

solution coated on a shaped metal pin by<br />

dipping it a into gelatin solution, IM can be<br />

used to prepare capsules, for example, the<br />

FlexTab TM technology that Capsugel acquired<br />

in 2011.<br />

More recently, IM has been used to<br />

directly incorporate APIs into shaped plastic<br />

parts, and hence used to prepare drug products.<br />

The majority of drug products prepared by IM<br />

are drug-eluting devices (DED); however, even<br />

more recently, IM has been used to prepare<br />

solid oral dosage forms (SOD). IM offers the<br />

product developer novel delivery features,<br />

specific shaped-part preparation capability, and<br />

potential for life-cycle management of APIs.<br />

Commercial DED prepared by IM include<br />

intravaginal rings (IVR), and several such<br />

devices on the market made of silicones<br />

manufactured using a RIM process. Examples<br />

of such IVR are FemRing ® , Estring ® , and<br />

Progering ® for hormone replacement therapy,<br />

vaginal atrophy, and contraception,<br />

respectively. These are core-sheath reservoir<br />

devices in which a drug-loaded silicone core is<br />

coated with a drug-free silicone sheath to<br />

regulate the rate of release of API from the<br />

device, yielding virtually zero-order (constant)<br />

release kinetics. The sheath is put over the core<br />

in a second injection molding process, making<br />

manufacturing quite complex.<br />

The International Partnership for<br />

Microbicides (IPM) working with Karl<br />

Malcolm and David Wolfson at Queens<br />

University, Belfast, has leveraged silicone<br />

technology in the development of a simpler<br />

IVR containing the non-nucleoside reverse<br />

transcriptase inhibitor dapivirine that does not<br />

have a rate controlling membrane. 1 This IVR is<br />

a device to protect women from HIV<br />

transmission during sexual intercourse with an<br />

infected partner, and is slated to start Phase III<br />

clinical trials in 2012.<br />

The RIM process requires the API and<br />

any other excipients to be suspended in the<br />

silicone liquids prior to injection (silicones are<br />

poor solvents so all added materials are<br />

suspended). Challenges arise from aggregation<br />

and settling of particulate materials in these<br />

fluids, which can cause inhomogeneities and<br />

nozzle blocking.<br />

Particle Sciences uses IM in the<br />

development of EVA and polyurethane DED<br />

for a variety of clients. 2,3 EVA and<br />

polyurethanes are thermoplastic polymers, and<br />

APIs and additives can be co-mixed uniformly<br />

with it prior to IM using hot-melt extrusion to<br />

yield pellets that are stable and can be used<br />

right away or stored for later IM processing.<br />

IVRs are developed first at laboratory scale<br />

using a bench-top molder, and successful<br />

formulations are then scaled to larger molding<br />

units for clinical and then commercial process<br />

development. The in vitro release of dapivirine<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

39


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

40<br />

Schematic of the Egalet process.<br />

from an EVA IVR made by molding<br />

dapivirine-loaded pellets (1.3% w/w API) in a<br />

mold with a torroidal cavity is shown in Figure<br />

4 orange markers), along with the drug-release<br />

profile fitted using a proprietary predictive<br />

model (blue line) showing excellent fit to the<br />

data. The diffusion coefficient of dapivirine in<br />

EVA can be determined from these plots,<br />

which in turn allows the prediction of release<br />

profiles from EVA IVRs of different strengths<br />

to be made.<br />

The shape of the curve is typical of<br />

release from monolithic devices that do not<br />

have release-rate controlling membrane, such<br />

as the aforementioned silicone IVR, according<br />

to Fick’s law for diffusion modified for the<br />

geometry of the part. 4 Such products are<br />

appropriate when a high release on day 1 of<br />

use compared to later time points is acceptable<br />

or desired.<br />

Very small shaped parts can be made by<br />

IM. For example, punctual plugs - small<br />

polymer pieces shaped to fit into the orifice<br />

through which tears drain (the puncta) - are<br />

used in the treatment of dry eye and other<br />

diseases of the eye. They can be pure polymer<br />

or medicated with API. In either case, they are<br />

prepared by micro-injection molding.<br />

Researchers at the University of Ghent in<br />

Belgium, have used IM to prepare API-loaded<br />

tablets for oral sustained release. 5 For example,<br />

metoprolol tartrate (MPT) was formulated into<br />

a mixture of polyethylene oxide (PEO) and<br />

ethyl cellulose (EC). The formulations were<br />

injection molded into biconvex tablets using a<br />

Haake MiniJet TM lab-scale injection molder,<br />

and the in vitro release profiles of the<br />

MPT/PEO/EC tablets were compared to<br />

compression molded tablets. The workers<br />

F I G U R E 5<br />

found a more sustained-release from the<br />

injection molded tablets.<br />

The Danish pharmaceutical company<br />

Egalet developed a two-step IM process to<br />

prepare oral dosage forms. The first step<br />

involves molding an open-ended “tube” of<br />

non-degradable polymer into which a<br />

thermoplastic API/polymer formulation is<br />

molded in a second step. 6 <strong>Drug</strong> is released by<br />

erosion from the open ends. The process is<br />

illustrated in Figure 5.<br />

SUMMARY<br />

Injection molding is a versatile process<br />

that has been in use in plastics processing for<br />

more than a century. More recently, it has been<br />

used in the pharmaceutical industry to prepare<br />

products from medical devices to complex<br />

controlled-release dosage forms for both oral<br />

and implantable routes of administration. u<br />

REFERENCES<br />

1. Malcolm RK, Edwards KL, Kiser P, Romano J, Smith TJ. Advances<br />

in microbicide vaginal rings. Antiviral Research. 2010;88[Suppl 1,<br />

S30-9].<br />

2. Clark MR, Kiser PF, Loxley A, McConville C, Malcolm RK,<br />

Friend DR. Pharmacokinetics of UC781-loaded intravaginal ring<br />

segments in rabbits: a comparison of polymer matrices. <strong>Drug</strong><br />

<strong>Delivery</strong> Translational Research. 2011;1(3):238-46.<br />

3. Loxley A, Gokhale A, Kim Y, McConnell J, Mitchnick M.<br />

Ethylene-vinylacetate intravaginal rings for zero-order release of an<br />

antiretroviral drug. Poster presented at the CRS annual meeting in<br />

NYC; 2008.<br />

4. Siepmann, Siepmann. Modeling of diffusion controlled drug<br />

delivery. J Controlled Release. (2011) in press.<br />

5. Quinten et al. <strong>Development</strong> and evaluation of injection-molded<br />

sustained-release tablets containing ethylcellulose and polyethylene<br />

oxide. <strong>Drug</strong> <strong>Development</strong> and Industrial Pharmacy.<br />

2011;37(2):149-159.<br />

6. See http://www.egalet.com/index.dsp?area=32.<br />

Pr<br />

B I O G R A P H I E S<br />

Dr. Andrew<br />

Loxley is<br />

Director of New<br />

Technologies at<br />

Particles Sciences<br />

Inc., a contract<br />

research<br />

organization in<br />

Bethlehem, PA,<br />

specializing in<br />

pharmaceutical formulation development. He<br />

leads a variety of projects, based on novel and<br />

proprietary nanotechnologies and combination<br />

devices, in fields from HIV vaccine and<br />

microbicide development, to gene-silencing<br />

SiRNA delivery. Prior to joining Particles<br />

Sciences, he led development efforts in nextgeneration<br />

lithium ion batteries at A123<br />

Systems Inc, electrophoretic displays at E<strong>IN</strong>K<br />

Corp., and emulsion polymers at Synthomer<br />

Ltd. British-born, he earned his BSc in<br />

Chemistry from the Univeristy of Sussex and<br />

his PhD in Physical Chemistry focusing on<br />

microencapsulation from the University of<br />

Bristol.<br />

Brett Braker is<br />

a Formulator at<br />

Particle Sciences,<br />

focusing on the<br />

development of<br />

drug-eluting<br />

devices. He earned<br />

his BS in Plastics<br />

and Polymer<br />

Engineering<br />

Technology from Pennsylvania College of<br />

Technology.


MATHEMATICAL<br />

M O D E L I N G<br />

Mathematical Modeling for Faster<br />

Autoinjector Design<br />

By: Jonathan Wilkins, PhD, and Iain Simpson, PhD<br />

<strong>IN</strong>TRODUCTION<br />

There is an increasing demand for<br />

advanced injection devices that bring<br />

benefits around self-administration and<br />

ease of use and work with new biological<br />

drugs, which are often required to be<br />

delivered in larger volumes and/or at<br />

higher viscosities than those for<br />

conventional drugs. In order to reduce<br />

development and manufacturing costs,<br />

there is also a desire for platform<br />

devices, which can meet a broader range<br />

of drug and user requirements through<br />

simple adaptation of a core design.<br />

Unfortunately, a number of devices<br />

currently in the market do not meet these<br />

requirements and furthermore, there is<br />

often a lack of a detailed understanding<br />

of how the key design parameters affect<br />

the overall device performance. In some<br />

cases, this has led to product recalls and<br />

in other cases, resulted in challenges in<br />

adapting the designs to meet new needs.<br />

Hence, there is a commercial need in the<br />

injectables industry for better<br />

understanding of how design<br />

fundamentals affect the performance of<br />

autoinjector devices.<br />

Cambridge Consultants believe a<br />

fast and effective approach to a better<br />

injection device design is a novel<br />

combination of mathematical modeling<br />

on a desktop PC, and complementary<br />

experimental data. The use of<br />

mathematical modeling gives insight into<br />

the physical behavior of the device, and<br />

allows rapid prediction of the effect of<br />

different parameters during the design<br />

process. The more established<br />

experimental approach provides<br />

confirmatory data to support the<br />

modeling. Cambridge Consultants has<br />

deployed this methodology in a number<br />

of development projects and achieved<br />

benefits in terms of reduced<br />

development time, more robust and<br />

Schematic of the Autoinjector<br />

F I G U R E 1<br />

adaptable designs, and reduced product<br />

cost.<br />

Mathematical modeling allows for<br />

quick simulations of device performance.<br />

The effect of important design<br />

parameters, such as injection time,<br />

injection force, and shear stress on the<br />

drug, can be predicted in real-time. This<br />

allows the engineer to investigate the<br />

design space and quickly optimize a<br />

suitable set of components for a new<br />

injection device. For example, the<br />

mathematical model can guide the<br />

designer in choosing the correct driving<br />

spring, needle gauge, and plunger in<br />

order to meet the device performance<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

41


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

42<br />

specification.<br />

In this paper we present a mathematical<br />

model of device performance applied to a<br />

typical autoinjector. This has the generic<br />

features of an autoinjector, but is not tied to<br />

any specific commercial device, and thus,<br />

can be tailored to investigate a wide range of<br />

interesting design features. The approach and<br />

the type of physics used inside the model are<br />

described. How experimental data can be<br />

used to calibrate and verify the model is also<br />

shown.<br />

As a case study, we use the<br />

mathematical model to predict the effect<br />

needle tolerances have on injection times<br />

within an autoinjector. This is an essential<br />

piece of performance data and is something<br />

that would take weeks and an expensive<br />

budget to determine experimentally, but that<br />

can be done in a few hours with a<br />

mathematical model.<br />

THE AUTO<strong>IN</strong>JECTOR MODEL<br />

The generic autoinjector shown in<br />

Figure 1 and considered for the model has<br />

the following components, which are<br />

commonly found in most commercial<br />

devices:<br />

• A syringe containing the drug<br />

• A needle<br />

• A compliant plunger to seal the drug<br />

into the syringe<br />

• A drive spring as the energy source<br />

that powers the plunger<br />

• A liquid drug<br />

• An air bubble between the drug and<br />

the plunger<br />

Easy-to-Use GUI for the Model<br />

The trigger that activates the<br />

autoinjector by releasing the spring force on<br />

to the plunger has not been considered. The<br />

physics of each component can be considered<br />

in isolation using ordinary differential<br />

equations that vary with time.<br />

The needle is modeled as a component<br />

that creates viscous pressure losses via the<br />

Hagen Poiseuille equation and inertial<br />

<strong>Drug</strong> Shear Strain Rate Predictions<br />

F I G U R E 2<br />

F I G U R E 3<br />

pressure losses at the entry and exit. The drug<br />

is modelled as a Newtonian fluid. The<br />

plunger is modelled as a rubber component<br />

with linear compliance, subject to a<br />

maximum compression limit. The syringe<br />

exerts a frictional force on the plunger. For<br />

the purpose of the model, a 1 ml BD Hypak<br />

syringe was used as a representative<br />

commercially available syringe. The air


Injection Time Sensitivities to Needle Tolerances<br />

bubble is modelled as a compliance, with an<br />

internal pressure related to the amount of<br />

compression via Boyle’s Law. The spring is<br />

modelled as a linear compression spring; it is<br />

initially compressed and expands during<br />

device activation. It would be a relatively<br />

minor change to the mathematical model to<br />

replace the mechanical spring with a motor<br />

energy source as used in electronic<br />

autoinjectors like the EasyPod.<br />

F I G U R E 4<br />

F I G U R E 5<br />

Experimental & Simulated Plunger Forces for a Verification Test Case<br />

The individual equations describing<br />

each component are assembled into a<br />

mathematical model of the entire autoinjector<br />

system, where all the components are<br />

interdependent. We use the Simulink<br />

commercial package to solve this system of<br />

equations. To make the model user friendly<br />

for a wide audience, we built a front-end<br />

GUI in Matlab. As shown in Figure 2, the<br />

GUI allows for key design parameters to be<br />

changed easily, has a graphical representation<br />

of the syringe plunger motion, and also plots<br />

out metrics of interest such as plunger<br />

motion, drug pressure, and percentage of<br />

drug delivered as a function of time.<br />

The model allows us to look at the<br />

physical behavior of the autoinjector at<br />

various points in time. Some of the<br />

parameters we predict are easy to calculate<br />

mathematically but difficult to monitor<br />

experimentally: for instance, shear stress on<br />

the drug. However, knowledge of the shear<br />

stress on the drug is important because high<br />

levels of shear stress can damage and<br />

degrade the molecules within the drug:<br />

particularly newer biologic drugs consisting<br />

of complex chains of proteins. An example<br />

shear stress simulation is shown in Figure 3<br />

for a drug with viscosity 6 times greater than<br />

water that is being driven by a spring with<br />

stiffness of 600 N/m with either a 27-gauge<br />

or a 25-gauge needle. Initially, the shear<br />

stress is several orders of magnitude higher<br />

than the steady state value. This is due to the<br />

impact of the initial spring release that causes<br />

rapid compression of the bubble, and<br />

subsequent high driving pressures and<br />

velocities for the drug in the needle. It is<br />

found that that the smaller diameter 27-gauge<br />

needle has a peak shear strain rate<br />

approximately 50% higher than for the 25<br />

gauge.<br />

The model also highlights the<br />

sensitivities in the autoinjector design. For<br />

example, consider the effect of needle<br />

diameter ±5% tolerances (nominally 27<br />

gauge) and needle length ±5% tolerances<br />

(nominally 0.5 inch) on the time to deliver 1<br />

ml of drug with a viscosity equivalent to<br />

water. Injection time is an important metric<br />

because the patient will prefer a shorter<br />

duration injection rather than a long one.<br />

The contour plot in Figure 4 predicts<br />

that injection time is much more sensitive to 43<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

needle diameter than to needle length. A<br />

difference of 23% in injection time results<br />

from changing the needle diameter by only -<br />

5% to +5% of nominal. This knowledge<br />

allows the designer to focus on the needle<br />

diameter as the single most powerful design<br />

parameter that can be used to adjust injection<br />

time.<br />

A designer may have to adjust the<br />

design to compensate for changes in the drug<br />

formulation. This is particularly true for<br />

platform devices in which a standard<br />

mechanical layout is customized to suit a<br />

range of different drugs. The mathematical<br />

model can be used to predict how the design<br />

must be altered to cope with a change in drug<br />

properties such as viscosity.<br />

For instance, suppose an initial version<br />

of the device platform is developed to work<br />

with a viscosity equivalent to water (1<br />

centiPoise) and has an injection time of 1.6<br />

seconds. This is achieved by using a spring of<br />

stiffness 600 N/m. If a new drug formulation<br />

has a viscosity that is five times higher (5<br />

centiPoise), and there is a requirement that<br />

the injection time should remain constant, the<br />

model predicts that to achieve this, a spring<br />

stiffness of approximately 1250 N/m will be<br />

now be required. This analysis takes minutes<br />

to perform with the mathematical model, but<br />

would require a lengthy program involving<br />

many different mechanical prototypes to do<br />

experimentally. Having determined the<br />

required higher spring force, the rest of the<br />

design can then be re-evaluated and modified<br />

if necessary to ensure it can function with the<br />

stiffer spring.<br />

EXPERIMENTAL VERIFICATION<br />

It is important to be able to confirm that<br />

the underlying equations representing each<br />

component in the model are realistic, and that<br />

44<br />

Monte Carlo Tolerance Analysis Predictions<br />

the output from the mathematical model can<br />

be trusted. A way to verify the model is to<br />

compare it with experimental data of the<br />

force required to drive the plunger on a<br />

representative commercially available<br />

syringe. The measurements were made on a<br />

Mecmesin force-displacement tester, which<br />

drives the syringe plunger at constant<br />

velocity and records the required force. This<br />

is a slightly different scenario from the<br />

original mathematical model in which a<br />

known force from the spring is applied and<br />

the velocity of the plunger is calculated.<br />

Thus, a modified version of the mathematical<br />

model was developed specifically for<br />

verification in which the velocity of the<br />

plunger is specified and the plunger force is<br />

calculated. The verification model was<br />

compared against various different<br />

experimental test cases. A sample<br />

verification of the plunger force<br />

mathematical model predictions and<br />

measurements is given in Figure 5 for a test<br />

case with a 2.5-mm long air bubble in a 1-ml<br />

syringe, a 30-gauge needle, and a test liquid<br />

six times more viscous than water.<br />

Predictions and measurements are in good<br />

agreement.<br />

F I G U R E 6<br />

CASE STUDY: MANUFACTUR<strong>IN</strong>G<br />

TOLERANCE ANALYSIS<br />

A common problem in injection device<br />

development is to understand the effects of<br />

manufacturing tolerances on the design. The<br />

designer must ensure the device performs<br />

within specification over the range of<br />

component tolerances, whilst considering that<br />

relying on excessively tight manufacturing<br />

tolerances can make a design uneconomic.<br />

We can apply the mathematical model to<br />

predict the sensitivities of an autoinjector to<br />

realistic manufacturing process tolerances to<br />

see whether a design will always perform to<br />

specification.<br />

For example, the mathematical model<br />

can be used to aid the designer in<br />

understanding how differences in spring<br />

stiffness or shape arising from production<br />

variations might affect drug delivery<br />

performance. For example, for a 5-centiPoise<br />

formulation, a 10% reduction in spring force<br />

from 1250 N/m increases injection time by a<br />

similar percentage, thus the dependence is<br />

not so critical.<br />

As another example, consider a device<br />

with a 1-ml BD HyPak syringe with a


nominal 27-gauge needle of 0.5-inch length,<br />

and investigate the effect of tolerances on<br />

injection time. We assume that each needle<br />

diameter, needle length, and syringe diameter<br />

vary with a process variation of Cp = 1.33.<br />

Using a Monte Carlo approach, we<br />

simulate a sample of 1000 syringes that<br />

embody this distribution of tolerances, and<br />

the results are shown in Figure 6. The<br />

mathematical model is run once for each of<br />

these syringes, and the injection times are<br />

predicted. This takes a matter of hours. Doing<br />

this experimentally would take weeks, and<br />

would require a lot of test specimens to be<br />

made at representative tolerances that would<br />

be expensive.<br />

This Monte Carlo approach is more<br />

realistic and offers a narrower range of<br />

injection times than a simpler worst case<br />

tolerance analysis in which all tolerances are<br />

assumed to be at the maximum permissible<br />

extremes. Our combination of the<br />

mathematical model with a Monte Carlo<br />

approach means the designer has a more<br />

realistic knowledge of the effects of<br />

tolerances and can make statements, such as<br />

“90% of all devices will have an injection<br />

time between 1.5 and 1.9 seconds.” That the<br />

real distribution of tolerances are narrower<br />

than the worst case values means the<br />

designer can achieve a given outcome and<br />

still specify less-restrictive tolerances in the<br />

manufacturing processes. This will result in<br />

considerable cost savings, particularly with<br />

an autoinjector that is likely to be made in<br />

quantities of millions per year.<br />

SUMMARY<br />

This article has introduced a<br />

mathematical model of an autoinjector. The<br />

fundamental approach considers the generic<br />

physical features of a device, and can be<br />

tailored to model specific commercial<br />

products as required. The model has been<br />

written in the commercial package Simulink<br />

to make it quick to adapt and to run.<br />

Simulink solves the detailed mathematics<br />

describing the physics of the device. To make<br />

the model intuitive and easy to use, we<br />

created a graphical user interface, which<br />

means it is accessible to designers who do<br />

not necessarily need to know the specifics of<br />

the physics behind the device.<br />

We have shown how the model can be<br />

used at the early design stage to define key<br />

components, such as the drive spring or<br />

needle in order to meet the product<br />

requirements specification. The model also<br />

facilitates platform solutions by allowing the<br />

designer to predict rapidly the changes<br />

needed in an existing device to meet a<br />

change in drug specification. Much of the<br />

behavior the model can predict is difficult to<br />

measure experimentally, such as shear stress<br />

in the drug or behavior over short timescales,<br />

but gives valuable insight into how the device<br />

functions. The model is also valuable during<br />

the manufacturing scale-up process, as it uses<br />

a Monte Carlo approach to simulate the<br />

effect of tolerance interactions on device<br />

performance. This is particularly expensive to<br />

do experimentally, as it would require a large<br />

range of samples to be manufactured and<br />

tested.<br />

We believe more extensive use of<br />

mathematical modelling in combination with<br />

experimental testing throughout the<br />

development process can lead to more robust<br />

platform injection devices hence reducing the<br />

risk of product recalls and allowing more<br />

reliable and cost-effective adaption of device<br />

designs for the delivery of new therapies. u<br />

Pr<br />

B I O G R A P H I E S<br />

Dr. Jonathan<br />

Wilkins led<br />

a range of<br />

drug delivery<br />

device<br />

developments<br />

as Senior<br />

Consultant at<br />

Cambridge<br />

Consultants:<br />

from inhalers<br />

through to novel injection systems. He<br />

has an interest in applying mathematical<br />

modelling techniques to speed up the<br />

development and optimization times of<br />

new products. He has an engineering<br />

degree and PhD from Imperial College,<br />

University of London, specializing in fluid<br />

mechanics. He is currently Qualification<br />

Manager at Magma Global, developing<br />

novel materials and processes for the oil<br />

and gas industry.<br />

Dr. Iain<br />

Simpson is<br />

Associate<br />

Director of<br />

<strong>Drug</strong> <strong>Delivery</strong><br />

in the Global<br />

Medtech<br />

Practice at<br />

Cambridge<br />

Consultants.<br />

He has a 20year<br />

track record of multidisciplinary<br />

technology and product development, the<br />

last 12 of which have been spent mainly<br />

in drug delivery covering parenteral,<br />

pulmonary, nasal drug delivery as well as<br />

more invasive device technology of which<br />

he has mainly worked in program<br />

management and review roles. He has<br />

written and presented papers on a range<br />

of drug delivery topics, including<br />

developing inhalers for children,<br />

technology licensing, improving device<br />

compliance, and usability. He also<br />

lectures on drug delivery to the<br />

Cambridge University Masters in<br />

Bioscience Programme and is a past<br />

Chairman of the R&D Society.<br />

Dr. Simpson can be reached at<br />

iain.simpson@cambridgeconsultants.com<br />

and to whom correspondence should be<br />

addressed.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

45


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

46<br />

PULMONARY<br />

D E L I V E R Y<br />

A Next-Generation Inhaled Dry Powder<br />

<strong>Delivery</strong> Platform<br />

By: Jean C. Sung, PhD<br />

PRESSURIZED METERED DOSE<br />

<strong>IN</strong>HALERS (PMDIS)<br />

Pressurized metered dose inhalers<br />

(pMDIs) contain drug suspended or<br />

dissolved in a volatile propellant that is<br />

atomized for inhalation. The propellants<br />

that were originally used,<br />

chlorofluorocarbons and<br />

hydrofluoroalkanes, emit environmentally<br />

unfriendly gases. Moving away from these<br />

propellants has proven to be difficult with<br />

certain drugs and formulations, reducing<br />

the number of therapeutics that can be<br />

formulated in this way. pMDIs are also<br />

characterized by low lung deposition<br />

efficiency and require breath coordination<br />

for effective delivery.<br />

<strong>IN</strong>TRODUCTION<br />

<strong>Drug</strong> developers have long sought effective pulmonary delivery of therapeutics to treat diseases inherent to the<br />

respiratory tract. Indeed, inhaled therapies are certainly favored over oral or intravenous therapies for respiratory and other<br />

diseases. Pulmonary drug delivery offers superior direct targeting to the site of action, higher lung doses, and lower systemic<br />

drug concentrations, opening the potential for higher therapeutic index with an overall benefit of lower total body dose and<br />

reduced potential for adverse events. 1 <strong>Delivery</strong> of drug to the deep lung can also offer improved access to the systemic<br />

circulation with several advantages, including rapid onset of action, avoidance of first-pass metabolism, and convenience as<br />

compared to injection.<br />

Traditional inhaled drug delivery has used a number of technologies, including pressurized metered dose inhalers<br />

(pMDIs), nebulizers, and dry powder inhalers (DPIs). While delivery of drugs via these first-generation inhaled drug systems<br />

provides great advantages over oral or intravenous delivery, these systems also have inherent limitations. The limitations of<br />

these systems create a tremendous opportunity for next-generation inhaled delivery platforms that can overcome the<br />

shortcomings of today’s approaches.<br />

NEBULIZERS<br />

Nebulizers deliver atomized aqueous<br />

drug solution by air jet or ultrasonic<br />

mechanisms. Nebulized drugs are typically<br />

delivered continuously over multiple<br />

breaths. Used mostly for elderly, infant, or<br />

critically ill patients, nebulization is<br />

typically considered to be a lessconvenient<br />

delivery system in terms of<br />

portability and delivery time, and is also<br />

characterized by low lung deposition<br />

efficiency. Nebulizers possess limitations<br />

in terms of the formulation of drugs that<br />

are degraded by shear and air-water<br />

interfaces.<br />

DRY POWDER <strong>IN</strong>HALERS<br />

(DPIS)<br />

Dry powder inhalers (DPIs) have<br />

traditionally used micronized powdered<br />

medication blended with a large quantity<br />

of lactose-based carrier, limiting the<br />

amount of drug that can be delivered. With<br />

no propellant, DPIs generally rely on the<br />

force of patient inhalation for delivery,<br />

which has limited their use in patient<br />

populations with potentially compromised<br />

lung capacity, such as children and the<br />

elderly. These lactose blends are typically<br />

composed of more than 80% to 90%<br />

lactose with microgram quantities of drug,<br />

resulting in a low drug mass-to-volume of<br />

powder ratio that limits their use primarily<br />

to high-potency drugs. These powders are<br />

also generally highly flow rate-dependent<br />

with respect to their dispersibility, have


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

48<br />

poor delivery efficiency with typically less than<br />

20% of drug making it to the lung, and have<br />

high patient-to-patient variability. Second-<br />

generation DP delivery based on particle<br />

engineering approaches (rather than active<br />

devices) have included production of porous<br />

particles and coating of particles with<br />

hydrophobic force-modifying excipients, such<br />

as magnesium stearate. Porous particles allow<br />

for aerosolizable powders with good<br />

dispersibility over a wide range of inspiratory<br />

flow rates, however, the inherent low particle<br />

density results in a low drug mass-to-volume of<br />

powder inhaled. The reduction in amount of<br />

drug per unit volume can make porous<br />

particles unsuitable for large molecule drugs or<br />

drug combinations that often require higher<br />

effective drug mass loadings per dose.<br />

Limitations of existing inhaled drug<br />

delivery methods have created the opportunity<br />

for a new approach to DP inhaled drug delivery<br />

technology. Moving forward, trends in the<br />

industry indicate that this significant<br />

opportunity is growing as more pharmaceutical<br />

and biopharmaceutical companies pursue the<br />

following product and formulation objectives:<br />

• using pulmonary delivery for a range of<br />

small-to-large drug molecules, spanning<br />

both existing and new drug entities;<br />

• increasing the dosage of active drug<br />

F I G U R E 1<br />

Scanning Electron Micrograph of iSPERSE Particles<br />

(scale bar presents 2 microns).<br />

molecules in inhaled therapeutics,<br />

specifically increasing the drug mass to<br />

inhaled volume;<br />

• seeking to commercialize drug<br />

combination formulations, including<br />

two, three, and higher numbers of drugs<br />

combined into a single inhaled product;<br />

and<br />

• pursuing reproducible delivery across a<br />

range of patient populations, including<br />

pediatrics, the elderly, and those with<br />

generally compromised lung function.<br />

A new dry powder formulation approach<br />

has been developed that overcomes the<br />

limitations of traditional inhaled delivery and<br />

has the potential to expand therapeutic options,<br />

disease targets, and patient populations for<br />

pulmonary drug delivery.<br />

NEW PLATFORM FOR DPI<br />

THERAPEUTICS ADDRESSES<br />

LIMITATIONS, BROADENS SCOPE<br />

Based on research to address the<br />

limitations of existing inhaled dry powder<br />

formulations, Pulmatrix has developed<br />

iSPERSE TM , a novel, proprietary inhaled dry<br />

powder delivery platform for use in the<br />

pulmonary delivery of drugs. iSPERSE<br />

particles can be engineered for localized<br />

therapeutic applications in the lungs, or for<br />

systemic delivery of therapeutics in which the<br />

goals include rapid onset of action, avoidance<br />

of first-pass metabolism, and convenience as<br />

compared to injection.<br />

iSPERSE powders are characterized by<br />

small particle size, relatively high density and<br />

flow rate-independent dispersibility, along with<br />

the ability for low or high drug loading of<br />

single or multiple drugs. iSPERSE represents a<br />

next-generation pulmonary delivery platform<br />

with significant potential as iSPERSE’s<br />

properties yield drug delivery capabilities<br />

superior to (and indeed not feasible with)<br />

conventional dry powder technologies that rely<br />

on the use of lactose blending or low-density,<br />

porous particles.<br />

Pulmatrix, a clinical-stage biotechnology<br />

company discovering and developing a new<br />

class of therapies for respiratory diseases,<br />

developed this new approach to inhaled drug<br />

delivery as part of the development process for<br />

the company’s own novel, proprietary inhaled<br />

medicines, some of which are already in<br />

human clinical trials for a range of diverse<br />

diseases, such as asthma and chronic<br />

obstructive pulmonary disease (COPD).<br />

CUSTOMIZABLE DRUG-TO-<br />

CARRIER VOLUME<br />

iSPERSE uses proprietary salt-based<br />

formulations optimized for inhalation to create<br />

a robust and flexible platform that can<br />

accommodate low or high drug loads of a<br />

range of molecule types. iSPERSE particles<br />

(Figure 1) are routinely prepared by a singlestep<br />

spray-drying process from either aqueous<br />

or organic systems. Small hydrophilic, small<br />

hydrophobic, and large molecules have all been<br />

incorporated successfully into iSPERSE<br />

formulations. These iSPERSE powders can<br />

contain as little as 5% excipients, which<br />

compares favorably to the greater than 80% to<br />

90% lactose that is typical of commercial<br />

lactose blend DPI formulations.<br />

This fundamental formulation difference<br />

of iSPERSE, along with the powder property<br />

of relatively high density, maps directly into<br />

drug dose, creating feasible drug doses in a


unit of up to 100 mg for iSPERSE. 2<br />

Additionally, iSPERSE inherently offers the<br />

potential of a strong safety profile, as, in<br />

addition to drug molecules, iSPERSE dry<br />

powders exclusively contain excipients that are<br />

generally regarded as safe (GRAS).<br />

DYNAMIC FLOW RATE EFFICACY<br />

iSPERSE powders allow for the highly<br />

efficient delivery of reproducible aerosols to<br />

the lungs with mass median aerodynamic<br />

diameters (MMAD) typically ranging from 2 to<br />

5 microns and respirable fine particle fractions<br />

routinely greater than 50%. The iSPERSE<br />

particles also possess the desirable property of<br />

being highly dispersible across a wide range of<br />

dispersion energies in spite of their small<br />

geometric particle size. Across flow rates from<br />

15 through 60 liters per minute (LPM), the<br />

percent of powder emitted from a passive dry<br />

powder inhaler (DPI) using iSPERSE<br />

formulated DPs is high and remains primarily<br />

unchanged (Figure 2), delivering drugs to the<br />

lung much more efficiently and with far less<br />

energy on the part of the patient.<br />

iSPERSE powders improve upon the<br />

delivery efficiency limitations of lactose<br />

blends, in particular allowing for a reduction in<br />

nominal dose. This expands the potential<br />

therapeutic applicability of iSPERSE as it<br />

could be appropriate for the broadest patient<br />

populations, including effective inhaled drug<br />

delivery to patients with normal or impaired<br />

lung function, using simple and convenient<br />

commercially available inhaler devices, such as<br />

passive capsule or blister-based DPI devices.<br />

Furthermore, iSPERSE formulations can be<br />

readily made in both clinical trial and<br />

commercial quantities using the proven and<br />

scalable spray-drying process capable of high<br />

and consistent yields.<br />

MULTI-DRUG COMB<strong>IN</strong>ATIONS<br />

The properties of iSPERSE have<br />

meaningful therapeutic and patient benefits,<br />

including the potential for single formulations<br />

that contain multiple drugs. In fact, preclinical<br />

data have shown the potential of the iSPERSE<br />

platform to enable the aerosol delivery of drug<br />

combinations that include triple drug<br />

combinations or higher. For example, in in vitro<br />

and preclinical studies presented recently, an<br />

iSPERSE fluticasone and salmeterol<br />

combination was matched to commercially<br />

available Advair ® Diskus ® , which contains the<br />

fluticasone and salmeterol combination<br />

blended with lactose to enable pulmonary<br />

delivery. 3 A triple iSPERSE combination of<br />

Advair components and an additional<br />

anticholinergic bronchodilator was also<br />

demonstrated. Highlights from these data<br />

include:<br />

• iSPERSE demonstrated improved<br />

delivery efficiency over Advair<br />

Diskus, as iSPERSE was shown to<br />

deliver over 2 times more lung<br />

dose of the active pharmaceutical<br />

ingredients than Advair Diskus.<br />

This improved delivery efficiency<br />

may offer two primary benefits:<br />

reduced off-target drug exposure<br />

and oral deposition, which<br />

potentially could reduce side<br />

effects such as thrush (oral<br />

candida) and other infections and<br />

reduced nominal dose (dose<br />

sparing), which lowers cost of<br />

goods.<br />

F I G U R E 2<br />

iSPERSE powders are relatively flow rate independent and possess properties suitable for aerosol<br />

delivery. Two different iSPERSE powder formulations (A, B) exhibited consistent iSPERSE properties of<br />

being geometrically small, dispersible, and aerodynamically suitable for lung delivery. Capsuleemitted<br />

powder mass (CEPM) and volume median diameter (VMD) of iSPERSE powders emitted from<br />

a RS01 DPI as a function of flow rate and measured by laser diffraction (mean ± SD; n = 4-5). 3<br />

• iSPERSE showed flow rate-independent<br />

performance in terms of dose and<br />

particle size distribution, which could<br />

enable iSPERSE applicability across a<br />

broad range of patient populations,<br />

expanding applications beyond patients<br />

with normal lung function to also<br />

include those having lower or impaired<br />

lung function, including pediatric,<br />

elderly, and those with compromised<br />

lung function.<br />

• The iSPERSE particle size distribution<br />

that would reach the lungs is<br />

consistent with an Advair Diskus<br />

particle size distribution (MMAD<br />

between 3.1 and 3.2 microns for<br />

iSPERSE comparable to 3.0 microns<br />

for Advair Diskus).<br />

• iSPERSE showed excellent agreement<br />

in size distribution for both drugs<br />

(fluticasone and salmeterol) and, even<br />

with the addition of a third drug,<br />

iSPERSE was able to maintain<br />

comparable size distribution, flow rate<br />

independence, and other powder<br />

properties desirable for inhaled delivery.<br />

• Consistent delivery of dual and triple<br />

combination components was achieved,<br />

with all components of iSPERSE in<br />

both dual and triple combinations<br />

retaining expected in vivo activity, as<br />

demonstrated by reduced lung<br />

inflammation and airway hyper-<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

49


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

50<br />

responsiveness in a murine model of<br />

allergic asthma.<br />

<strong>IN</strong>HALED DELIVERY OF DIVERSE<br />

CLASSES OF DRUGS<br />

The potential of iSPERSE technology has<br />

been validated not only with small molecule<br />

drugs, but also macromolecule drugs (proteins,<br />

peptides, antibodies) at therapeutically relevant<br />

doses well in excess of those achievable by<br />

traditional dry powder lactose blend<br />

technologies. In vivo efficacy has been<br />

demonstrated with small molecules for the<br />

treatment of asthma and COPD, antibiotics in<br />

mouse models of bacterial infection, as well as<br />

lung and systemic delivery of macromolecules.<br />

POTENTIAL APPLICATIONS<br />

FOR ISPERSE<br />

The attributes of iSPERSE give this<br />

proprietary novel delivery platform the potential<br />

to (1) deliver high drug payloads, (2) deliver low<br />

potency drugs, (3) offer flexible formulation<br />

options, (4) reduce side effects, (5) facilitate<br />

straightforward manufacturing, (6) support the<br />

formulation of small and large molecule drugs<br />

(proteins and peptides), and (7) support drug<br />

combinations (including triple drug<br />

combinations or higher).<br />

Expanding the viability of dry inhaled<br />

powder delivery beyond select small molecules<br />

to include proteins, peptides, and antibodies at<br />

therapeutically relevant doses as well as triple,<br />

quadruple, or higher drug combinations will<br />

enable development of simple, convenient<br />

inhaled therapies for a new and expanded range<br />

of disease targets and patient populations. As<br />

such, iSPERSE has the potential to be the<br />

pulmonary delivery platform of choice for a<br />

number of first-in-class and best-in-class inhaled<br />

therapeutics.<br />

Commercially, Pulmatrix is seeking<br />

iSPERSE partnerships as well as advancing its<br />

own iSPERSE-based drug formulations. In<br />

terms of diseases that are likely near-term<br />

candidates for iSPERSE clinical initiatives, a<br />

number of proprietary iSPERSE drug<br />

formulation candidates are now being advanced,<br />

including small molecules, combinations, and<br />

biologics in a variety of therapeutic areas,<br />

including COPD, cystic fibrosis, asthma,<br />

idiopathic pulmonary fibrosis (IPF), and non-CF<br />

bronchiectasis.<br />

To support the development of its own<br />

pipeline as well as the iSPERSE partnering<br />

programs, Pulmatrix has developed a complete<br />

range of pulmonary drug formulation<br />

capabilities that are integral to the successful<br />

commercialization of the iSPERSE platform,<br />

including:<br />

• dry powder formulation and<br />

manufacturing,<br />

• dry powder physicochemical properties<br />

optimization,<br />

• aerosol characterization and method<br />

development,<br />

• dry powder inhaler selection and testing,<br />

• preclinical efficacy/safety testing (in<br />

vitro and in vivo), and<br />

• clinical program operation and<br />

management.<br />

These platform and formulation<br />

optimization capabilities will be offered to<br />

iSPERSE partners. u<br />

REFERENCES<br />

1. Patton and Byron, Inhaling medicines: delivering drugs to<br />

the body through the lungs. Nature Reviews <strong>Drug</strong><br />

Discovery, 2007;6:67-74.<br />

2. Sung et al. iSPERSE TM : formulation and in vitro<br />

characterization of a novel dry powder drug delivery<br />

technology. Respiratory <strong>Drug</strong> <strong>Delivery</strong> Europe. 2011;2:411-<br />

414.<br />

3. Sung et al. Pulmonary <strong>Delivery</strong> of Combination <strong>Drug</strong><br />

Products via a Novel Dry Powder <strong>Delivery</strong> Technology.<br />

Paper presented at the 11th US-Japan Symposium on <strong>Drug</strong><br />

<strong>Delivery</strong> Systems. Lahaina, Hawaii, December 15-20, 2011.<br />

Pr<br />

B I O G R A P H Y<br />

Dr. Jean C. Sung is the Director of<br />

Pharmaceutical <strong>Development</strong> at Pulmatrix, a<br />

clinical-stage company discovering and<br />

advancing novel respiratory therapeutics and<br />

drug delivery technologies. She is responsible<br />

for Pulmatrix’s formulation, process, and<br />

analytical development functions. With prior<br />

experience at Alkermes, Inc. and AIR, Inc.<br />

(Advanced Inhalation Research), Dr. Sung has<br />

spent more than a decade developing novel<br />

particle engineering and formulation<br />

technologies to advance respiratory dry<br />

powder drug delivery. Dr. Sung earned her PhD<br />

and MS in Engineering Sciences with a focus<br />

on Biomedical Engineering from Harvard<br />

University and her SB in Chemical Engineering<br />

from Massachusetts Institute of Technology.


SPECIAL FEATURE<br />

Transdermal, Topical &<br />

Subcutaneous: Non-Invasive <strong>Delivery</strong><br />

to Expand Product Line Extensions<br />

The overall landscape of the transdermal, topical, and subcutaneous markets continue to change - for the better - as non-invasive<br />

delivery of drugs to the skin or to the systemic circulation via the skin is a highly attractive proposition for many active drugs, be they<br />

poorly soluble or subject of first-pass metabolism when administered orally. Equally attractive is the possibility of administering<br />

chronic therapies via the skin, translating to convenience, safety, patience compliance, and drug efficacy. Thus, the pharmaceutical industry has<br />

an opportunity to expand on product line extensions for existing drugs.<br />

By: Cindy H. Dubin, Contributor<br />

In this <strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> feature, delivery system providers and contract developers and manufacturers were asked to<br />

describe their products and service offerings in their respective area of expertise.<br />

TRANSDERMAL DRUG DELIVERY<br />

The transdermal delivery market was valued at $21.5 billion in 2010 and is predicted to reach $31.5 billion by 2015, according to a<br />

PharmaLive Report. The annual US market for transdermal patches is estimated at more than $3 billion, and transdermal drugs account for<br />

more than 12% of the global drug delivery market. Transdermal drug delivery prevents many of the problems associated with oral and<br />

intravenous routes. Major advantages provided by transdermal drug delivery include improved bioavailability, more uniform plasma levels,<br />

F I G U R E 1<br />

3M <strong>Drug</strong> <strong>Delivery</strong> Systems develops a<br />

formulation of a drug-in-adhesive (DIA)<br />

patch product that delivers the drug<br />

through the skin in the targeted<br />

therapeutic dose range.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

51


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

52<br />

longer duration of action resulting in<br />

less dosing frequency, reduced side<br />

effects, and improved therapy due to<br />

maintenance of plasma levels.<br />

Transdermal drug delivery is used in<br />

areas such as pain management and<br />

women’s health. Pharmaceutical,<br />

biotechnology, and drug delivery<br />

companies are poised to tap the hidden<br />

potential in transdermal drug delivery<br />

applications, such as wound care,<br />

monitoring, and diagnostic methods.<br />

The development of successful<br />

transdermal drug delivery systems in<br />

these areas will play an important role<br />

in improving patient quality of life.<br />

3M <strong>Drug</strong> <strong>Delivery</strong><br />

Systems–Putting<br />

Convenience &<br />

Compliance Into a Patch<br />

3M <strong>Drug</strong> <strong>Delivery</strong> Systems serves as a<br />

contract manufacturer to pharmaceutical<br />

companies that market transdermal products,<br />

and also helps companies develop and adapt<br />

their drugs for transdermal delivery. 3M<br />

<strong>Drug</strong> <strong>Delivery</strong> Systems provides<br />

development services - taking drug products<br />

and adapting them into the transdermal<br />

dosage form. 3M <strong>Drug</strong> <strong>Delivery</strong> Systems can<br />

also provide basic contract manufacturing<br />

services by taking an existing product or one<br />

that it developed and manufacture it for the<br />

customer.<br />

“Clients interested in developing new<br />

drugs into transdermal products are often<br />

challenged with being able to achieve<br />

consistent delivery at the desired rate and<br />

pharmacokinetic profile to obtain the desired<br />

therapeutic effect,” says Jordan Fineberg,<br />

Global Business Manager, Transdermal <strong>Drug</strong><br />

<strong>Delivery</strong> Systems for 3M <strong>Drug</strong> <strong>Delivery</strong><br />

Systems. “Fortunately, we are able to provide<br />

very strong technical capabilities and<br />

analytics in our labs to help clients overcome<br />

those technical challenges.”<br />

To create a passive transdermal product,<br />

3M <strong>Drug</strong> <strong>Delivery</strong> Systems develops a<br />

F I G U R E 2<br />

formulation of a drug-in-adhesive (DIA) patch<br />

product that delivers the drug through the skin<br />

in the targeted therapeutic dose range.<br />

“Our DIA systems are versatile enough<br />

to be compatible with a variety of drugs, and<br />

our targeted market is companies with APIs<br />

that fit the transdermal profile. We help our<br />

customers develop their products in a way<br />

that achieves a consistent, controlled-release<br />

dosage level that can be difficult to achieve<br />

by other forms of administration,” adds Mr.<br />

Fineberg.<br />

Transdermal patches are distinguished<br />

by other systems by the duration for which<br />

they can be worn. Products can be created<br />

that deliver multi-day dosing at a very<br />

specific PK profile. Additionally, continued<br />

advancements allow this system to deliver<br />

more than one drug in a single patch, adding<br />

to convenience for patients. With the ability<br />

to deliver a controlled release of API over<br />

multiple days, patient compliance is possibly<br />

improved.<br />

“We see continued generic entrants into<br />

the transdermal market, particularly with a<br />

couple of big drugs that will go generic in<br />

the next 3 to 5 years,” predicts Mr. Fineberg.<br />

“In our own future, we see 3M <strong>Drug</strong><br />

Adhesives Research’s technology is a tailorable,<br />

pharmaceutical-grade acrylic adhesive that<br />

secures a patch/device for up to 7 days.<br />

<strong>Delivery</strong> Systems working with both branded<br />

and generic companies in developing and<br />

manufacturing their transdermal products.”<br />

Adhesives Research,<br />

Inc.–Responding to Skin-<br />

Friendly, Long-Term Wear<br />

Adhesives<br />

Adhesives Research specializes in the<br />

custom development of unique component<br />

adhesives, films, and laminates for<br />

transdermal and buccal drug delivery. The<br />

company offers complete customization of<br />

our platform technologies (examples such as<br />

wear-times, moisture vapor transmission,<br />

shear, etc.) to create the tailored functionality<br />

required of the adhesives used in a<br />

customer’s applications.<br />

Today, many companies are developing<br />

drug delivery devices that will be bonded to<br />

the skin for longer periods of time. The<br />

majority of transdermal patches and drug<br />

delivery devices available today are dailywear<br />

devices that are typically removed<br />

within 24 hours of application; however,<br />

formulators are developing extended-wear<br />

patches designed to be worn for multiple<br />

days, up to a week. In the case of insulin


infusion pumps, the adhesive must reliably<br />

attach a device to skin while bearing load.<br />

“While aggressive adhesion ensures a<br />

secure bond to skin for addressing dosing<br />

concerns, it can potentially cause discomfort<br />

upon patch removal,” says Mary Lawson,<br />

Pharmaceutical Business Manager, Adhesives<br />

Research, Inc. “Pain is caused when the<br />

adhesive removes skin cells and/or hair when<br />

the device is pulled away. Also, an aggressive<br />

adhesive that releases uncleanly may leave<br />

behind an unwanted residue on the skin that<br />

is difficult to remove.”<br />

In response to the need for skin-friendly,<br />

long-term wear adhesives, Adhesives<br />

Research’s newest technology addresses the<br />

need through a tailorable, pharmaceutical-<br />

grade acrylic adhesive technology that meets<br />

the critical design parameters for securing a<br />

patch/device for periods up to 7 days to<br />

ensure adequate skin bonding with residue-<br />

free removal.<br />

“In spite of the adhesive’s aggressive<br />

nature, the pain experienced upon removal is<br />

considered to be low-to-moderate, and<br />

studies have shown that removal of the<br />

adhesive tape does not cause disruption of<br />

the stratum corneum,” says Ms. Lawson.<br />

As a general rule, adhesives for<br />

F I G U R E 3<br />

DBV uses two proprietary manufacturing processes to load active dry compounds onto a polymeric<br />

film backing.<br />

transdermal patches are formulated to present<br />

aggressive bonds with flexibility and<br />

conformability to ensure the patch or device<br />

remains firmly in place without lifting or<br />

fall-off to ensure a therapeutic dose.<br />

Ms. Lawson explains, “Our long-term<br />

wear adhesives are designed to withstand<br />

physical activity, constant friction from<br />

clothing, periodic moisture exposure, and<br />

varying degrees of skin porosity and oil<br />

levels without shifting or moving.”<br />

Adhesives Research's work is<br />

complemented by its relationship with its<br />

subsidiary, ARx LLC, a collaborative<br />

technology partner. ARx LLC is experienced<br />

in developing polymer chemistries, that when<br />

combined with its client’s APIs and<br />

complementary materials, transform into<br />

transdermal, transmucosal, and soluble film<br />

products. This includes single- and multi-<br />

drug products as well as immediate- and<br />

controlled-release offerings. ARx LLC<br />

develops and manufactures innovative<br />

pharmaceutical products with a focus on<br />

unique technologies in oral, topical, and<br />

transdermal drug delivery. The ARx products<br />

range from first-in-class immediate-release<br />

OTC and prescription soluble films to multi-<br />

day, sustained-release transdermal patch<br />

delivery. The ARx sole objective is to work<br />

hand-in-hand with its pharmaceutical<br />

partners to develop and launch<br />

therapeutically relevant products that have the<br />

potential to extend a molecule's product<br />

lifecycle, deliver new compounds, or provide<br />

better product performance in the areas of<br />

bioavailability, compliance, and/or cost.<br />

“The model is evolving in how<br />

companies talk to us about their product<br />

design. A decade ago, our partners came to<br />

us at the onset of a project with a<br />

predetermined path for integrating a molecule<br />

into a delivery platform technology,” says<br />

Megan Greth, Marketing Manager II, ARx,<br />

LLC. “Today, the conversation is much more<br />

open from the start of a project and is first<br />

focused on understanding the improved<br />

therapeutic outcome followed by evaluating<br />

the various technologies available for<br />

delivery. Basically, the conversation has<br />

evolved to a broader dialogue in which we<br />

evaluate the best delivery options based on<br />

the patient population we are targeting.<br />

“The diversity of the Adhesives<br />

Research and ARx technology platforms<br />

inherently enables us to provide design<br />

F I G U R E 4<br />

A Dow scientist observing the mixing of a<br />

topical product and checking for consistency.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

53


flexibility and developmental support to our<br />

clients from concept to commercialization,”<br />

Ms. Greth concludes.<br />

DBV Technologies–Addressing<br />

Unmet Allergy Treatments for<br />

Young Children<br />

According to the World Health<br />

Organization, allergies constitute the world’s<br />

fourth largest public health issue and affect<br />

500 million people worldwide. Allegies affect<br />

somewhere between 25% and 40% of the<br />

adult population and more than 50% of<br />

children in developed countries.<br />

Environmental, pollution, and changing food<br />

habits are contributors to the rapid increase in<br />

the prevalence of allergies.<br />

DBV Technologies focuses on food and<br />

pediatric allergy desensitization. The<br />

company has developed a patented skin<br />

patch, Viaskin ® that puts the allergen into<br />

contact with the immune system via immune<br />

cells present in the superficial layers of the<br />

skin, while avoiding disruption of the<br />

blood/skin barrier. This approach, which<br />

significantly reduces the risk of serious<br />

anaphylactic shock, enables Viaskin to fulfill<br />

at least two critically important medical<br />

needs that were previously unmet, explains<br />

Pierre-Henri Benhamou, CEO of DBV. First,<br />

Viaskin targets food allergies, in which the<br />

risk of anaphylactic shock is high, and<br />

second, the Viaskin patch is suitable for<br />

desensitizing children under the age of 5.<br />

“Therefore, we are in a position to<br />

address allergies that are not treatable by<br />

conventional pharmaceutical therapies, he<br />

says.”<br />

When Viaskin, containing a specific<br />

allergen, is applied on the skin, the allergens<br />

are desposited locally on the skin and are<br />

taken up by the skin’s immune-competent<br />

cells, triggering the immune response. The<br />

allergens are taken up in the superficial<br />

layers of the skin without crossing the basal<br />

membrane and preventing the allergen from<br />

54<br />

reaching the bloodstream.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

DBV uses two proprietary<br />

manufacturing processes to load active dry<br />

compounds onto a polymeric film backing<br />

via electrostatic force: static powder and<br />

electrospray deposit, a precise method of<br />

layering a controlling solution of the allergen<br />

on the patch so that it is dry and stable.<br />

DBV has developed Viaskin Peanut for<br />

peanut allergy, Vaskin Milk for treating IgEmediated<br />

cow’s milk allergy, and Viaskin<br />

House Dust Mites for preventing asthma in<br />

young children. In December 2011, DBV<br />

received Fast Track Designation for its<br />

Viaskin Peanut clinical development<br />

program.<br />

“This designation was very important<br />

for us as we believe this translates to a real<br />

unmet medical need that patients, regulators,<br />

and DBV are willing to fulfill,” says Mr.<br />

Benhamou.<br />

TOPICAL DRUG DELIVERY<br />

Scientific and technological advances in<br />

recent decades have widened the scope of<br />

possibilities for therapies to or via the skin,<br />

enabling targeted delivery of drugs to<br />

specific layers of the skin, muscle tissue, or<br />

even to the systemic circulation with<br />

unprecedented precision and exactitude.<br />

Since the advent of the first transdermal<br />

F I G U R E 5<br />

Ei Inc. concentrates on developing topical medicines.<br />

patch in the late 1970s, the pharmaceutical<br />

industry has witnessed successful delivery of<br />

chronic therapies like hormones, nicotine,<br />

and NSAIDS systemically via the skin. These<br />

innovations have significant advantages over<br />

the oral route: patient compliance, improved<br />

plasma levels, and reduced side effects.<br />

Topical OTC drugs are an important market<br />

segment with forecast worth expected to<br />

exceed $870 million in 2014, according to<br />

Datamonitor. Antiseptic cleansers account for<br />

the largest part of the topical OTC medicines<br />

market at almost 60%, with anti-itch products<br />

in second place at almost 28%, followed by<br />

anesthetic products, which represent more<br />

than 10% of the segment.<br />

Dow Pharmaceutical<br />

Sciences–A One-Stop-Shop<br />

Approach to Topical<br />

<strong>Development</strong><br />

Dow Pharmaceutical Sciences provides<br />

contract development services and clinical<br />

manufacturing, both sterile and non-sterile, to<br />

the pharmaceutical and biotechnology<br />

industries. With a focus being solely on<br />

topicals, Dow Pharmaceutical Sciences<br />

spends a lot of time developing ophthalmic<br />

products. Its newest service, Sterile Product<br />

<strong>Development</strong>, supports the development of<br />

customers’ topical products containing NCEs,<br />

NMEs, or approved molecules, through to


GMP manufacturing of supplies for Phase I<br />

and Phase II clinical trials. Clients can access<br />

all services required to support development<br />

of their sterile product. These include<br />

formulation development and in vitro drug<br />

penetration testing, analytical method<br />

development, stability testing, GMP & GLP<br />

manufacturing, labeling, and distribution of<br />

clinical supplies to test sites worldwide. The<br />

company provides a full team of specialists<br />

focused solely on topical product<br />

development working in one location.<br />

“Our new One-Stop-Shop provides<br />

customers with their best chance of success<br />

and fastest route to market, saving them both<br />

time and money,” says Vanlee F. Waters,<br />

Assistant Director of Marketing, Dow<br />

Pharmaceutical Sciences.<br />

Dow’s turnkey development process<br />

includes preformulation (drug solubility,<br />

excipient, and solvent compatibility)<br />

evaluations, formula design, and development<br />

of multiple prototypes. Multiple prototypes<br />

based on different delivery systems are<br />

developed to maximize success for chemical<br />

and physical stability, release from the<br />

formulation, and penetration into the skin or<br />

tissue as desired.<br />

Mr. Waters says that Dow has experience<br />

in working with the FDA’s Dermatology<br />

division. “Working closely with the FDA<br />

F I G U R E 6<br />

In addition to excipient safety and functionality, characterization of topical formulations for<br />

sensory attributes is an area in which Gattefossé has expertise.<br />

provides us with an intimate knowledge of<br />

their Non-Clinical and Clinical requirements,<br />

allowing us to get more products approved for<br />

our customers. Our track record remains<br />

second to none: 31 New <strong>Drug</strong> Approvals for<br />

prescription topical dermatological products<br />

were approved by the FDA during 2005-11:<br />

10 of these formulations were developed at<br />

Dow,” he says.<br />

Going forward, Mr. Waters says Dow<br />

will stay the course long-term and continue to<br />

leverage its focus and experience in topical<br />

product development.<br />

“As a boutique CDO provider to large<br />

and specialty pharma, we will continue to<br />

leverage our expertise in topical product<br />

development, non-clinical, clinical, and<br />

regulatory affairs.”<br />

Ei Inc.–Laser Focused on<br />

Topicals<br />

Ei Inc. is focused solely on topical semi-<br />

solids and liquids, and it is this expertise that<br />

Roger Martin, Senior Vice President of Sales<br />

and Marketing, says makes the company<br />

stand out in a market that seems to be<br />

fragmenting into specialty fields.<br />

“Clients are not really looking for one<br />

provider to manufacture all of their tablets,<br />

injectibles, and topicals under one roof. They<br />

instead want a vendor who is an absolute<br />

expert at a given dosage form,” he says.<br />

Clients coming to Ei Inc. are looking for<br />

topical products, and thus aesthetics and<br />

stability of the formulation are both very<br />

important. “Our formulation scientists have<br />

decades of experience working with various<br />

APIs used in topical medicines, and they<br />

work diligently to create both an efficacious<br />

and pleasant delivery vehicle,” says Mr.<br />

Martin.<br />

Critical to Ei’s mission is to protect the<br />

client’s brand by offering a level of quality<br />

that never puts their product in jeopardy, and<br />

service levels that allow them to plan their<br />

business effectively, Mr. Martin stresses. “By<br />

offering complete development, analytical,<br />

stability, and manufacturing services for<br />

topical products, Ei has positioned itself to<br />

serve customers in this niche better than any<br />

other.”<br />

Ei recently announced its partnership<br />

with Keranetics in which Ei will start<br />

manufacturing the API for Keranetic’s wound<br />

care and burn care products. This is Ei’s first<br />

venture into API manufacturing.<br />

“Ei is laser focused on the topical<br />

marketplace, and we have expanded our<br />

R&D, analytical, and manufacturing<br />

capabilities to meet this demand,” explains<br />

Mr. Martin. “We have heavily invested in both<br />

people and equipment so that our services can<br />

meet or exceed the expectations of our<br />

customers.”<br />

Gattefossé–Excipients for Safe<br />

and Effective Topical <strong>Delivery</strong><br />

Gattefossé specializes in topical<br />

formulations by virtue of the products in its<br />

offering. Gattefossé designs, manufactures,<br />

and markets functional excipients globally.<br />

Each product is developed to meet a unique<br />

formulation challenge and is supported by<br />

research and data generated internally. Before<br />

launching, every excipient is tested to ensure<br />

a high safety profile and conformity to NF,<br />

EP, JP, and other well-recognized<br />

pharmacopoeia. Since 2004, for example, the<br />

company has been responsible for the<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

55


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

completion of more than 25 NF and EP<br />

monographs.<br />

Gattefossé has a range of solubilizers<br />

and penetration or permeation enhancers. For<br />

instance, many marketed products already<br />

include Transcutol ® , an excipient that<br />

facilitates passage of drugs across the stratum<br />

corneum, but creates a drug depot effect in<br />

subcutaneous layers of the epidermis.<br />

The company has a range of emulsifiers<br />

for one pot, cold process, and low-energy<br />

emulsification processes, also coded in<br />

innumerable products worldwide. The list<br />

includes Tefose ® 63, a self-emulsifying base,<br />

known for its innocuity and safety profile for<br />

mucosal/vaginal delivery of antifungals.<br />

Other products like Labrafil ® , Labrasol ® , and<br />

Capryol TM help modulate permeation across<br />

the epidermis.<br />

In 2011, Gattefossé announced the<br />

inauguration of a new research center in<br />

Saint-Priest, France. Bringing together the<br />

formulation and R&D laboratories,<br />

the18,000-sq-ft facility is built to encourage<br />

and facilitate exchange of ideas and resources<br />

between various groups engaged in excipient<br />

characterization and expansion of that<br />

knowledge in relation to functionality in<br />

different drug delivery systems.<br />

“Parallel to safety and regulatory<br />

qualifications, Gattefossé’s business culture<br />

places emphasis on understanding the<br />

specific characteristics and functionality of<br />

excipients in every formulation design.<br />

Developing and sharing that knowledge, as to<br />

how our excipients can help improve,<br />

differentiate, and innovate customers’<br />

products, is the key to the continued success<br />

of the company,” says Jasmine Musakhanian,<br />

Marketing & Scientific Director,<br />

Pharmaceutical Division, Gattefossé USA.<br />

Characterization of topical formulations<br />

for sensory attributes (feel, touch,<br />

spreadability, etc.) is another area in which<br />

Gattefossé has expertise. “The subject has<br />

traditionally been of interest to personal care<br />

customers, but is now expanding to<br />

56<br />

pharmaceutical preparations,” Ms.<br />

Musakhanian adds.<br />

Selecting the right excipient(s) for the<br />

intended delivery system, relative to the type<br />

of active drug entity and its targeted therapy<br />

is likely to be the key step in saving time and<br />

cost to market. Gattefossé assists customers<br />

with the selection process by providing<br />

pertinent data on physical-chemical<br />

information and formulation support.<br />

Skinvisible Pharmaceuticals,<br />

Inc.–Delivering Tailored<br />

Molecules<br />

Skinvisible is an R&D company that<br />

formulates new products and provides life<br />

cycle management by revitalizing products<br />

coming off patent with its patented, targeted<br />

drug delivery system called Invisicare®.<br />

Invisicare delivers drugs on, in, or through<br />

the skin with a controlled release and can be<br />

tailored to almost any type of molecule.<br />

Invisicare has multiple applications,<br />

including topical, transdermal, and mucosal<br />

(in development). Invisicare is a filmforming<br />

complex of hydrophilic and<br />

hydrophobic polymers that are readily<br />

available and used in the marketplace.<br />

“Our technology is not encapsulation, it<br />

is a simple manufacturing process with no<br />

special equipment and no shearing required.<br />

It is very compatible (flexible) with both<br />

water-insoluble and certain cationic active<br />

ingredients (positive, negative, and neutral)<br />

and certain water-soluble actives,” explains<br />

Doreen McMorran, Vice President Business<br />

<strong>Development</strong> and Marketing for Skinvisible.<br />

The formulations do not use alcohol, waxes,<br />

or other organic solvents and can be<br />

formulated into creams, lotions, sprays, or<br />

gels.<br />

Invisicare represents a “family” of<br />

Invisicare structures, each specifically<br />

formulated for specific needs. This includes<br />

increasing the release of actives (ie, a 3%<br />

imiquimod formulation can release 30% of<br />

the active), binding of products (for use for<br />

F I G U R E 7<br />

Skinvisible formulations can be formulated<br />

into creams, lotions, sprays, or gels.<br />

hand sanitizers, sunscreens, and sunless<br />

tanning products), and/or photostability of<br />

avobenzone for 8 hours.<br />

In 2011, Skinvisible was granted two<br />

patents: a sunscreen avobenzone<br />

photostability patent for the US and a<br />

technology patent for Europe. Skinvisible<br />

now has 40 patented formulations with<br />

various indications, all available for licensing<br />

on an exclusive basis, including a recently<br />

developed formulation for Netherton<br />

syndrome for which the company is seeking<br />

orphan drug status in the US and Europe.<br />

SUBCUTANEOUS DRUG<br />

DELIVERY<br />

Providing innovative, patient friendly<br />

drug delivery devices is increasingly<br />

becoming a requirement, not an option, when<br />

introducing therapeutic products involving<br />

traditional needle and syringe use as<br />

evidenced by the prevalence of autoinjections,<br />

prefilled syringes, and pressure jet<br />

applicators offered by most large<br />

pharmaceutical companies today.


F I G U R E 8<br />

Zosano’s ZP Patch Technology is a needlefree<br />

delivery system consisting of a patch<br />

applied by a reusable applicator.<br />

Zosano Pharma–Using a Patch<br />

to Overcome the Needle<br />

Experience<br />

Although prefilled syringes, auto<br />

injectors, and pressure jet applicators<br />

products have improved patient convenience<br />

and compliance by reducing preparation and<br />

delivery steps, many patients still associate<br />

them with a needle experience. Zosano<br />

Pharma aims for its ZP Patch (formerly<br />

known as Macroflux) to replace the syringe<br />

altogether in target compound areas in which<br />

the patient needs are the highest.<br />

Zosano’s ZP Patch Technology is a userfriendly,<br />

simple, and needle-free transdermal<br />

delivery system consisting of a patch applied<br />

by either a reusable applicator for therapies<br />

requiring daily chronic administrations or a<br />

disposable, single-use patch applicator<br />

system for acute or short-term applications.<br />

The ZP patch technology delivers therapeutic<br />

compounds via drug-coated microneedles on<br />

the patch that permeate the skin’s outer layer,<br />

provide rapid and efficient systemic delivery,<br />

ensure significant therapeutic effect - and is<br />

painless. Dry-coated drug on the thin<br />

microneedle patch allows for rapid delivery<br />

into the skin. The creation of pathways<br />

through the skin improves control of drug<br />

distribution throughout the patch treatment<br />

area, reduces the potential for skin irritation,<br />

and provides for efficient drug delivery and<br />

absorption.<br />

The ZP patch has proven capable of<br />

delivering a broad range of compounds,<br />

including peptides, proteins, small<br />

molecules, and vaccines. It offers the<br />

reliability and predictability associated with<br />

a “one-step” application without relying on a<br />

liquid injection or pre-treatment<br />

(permeation) in preparation for transdermal<br />

delivery.<br />

Zosano Pharma has identified 10 key<br />

drugs that best fit the criteria for the initial<br />

product portfolio of the ZP patch. The<br />

combined market for this portfolio is at least<br />

$20 to $30 billion dollars in worldwide<br />

sales. “Current efforts to increase our patch<br />

dosage delivery dovetails well with the need<br />

for product differentiation in the expanding<br />

biosimiliars market and will potentially<br />

increase the market estimate even further,”<br />

says Brian Rippie, Director of Business<br />

<strong>Development</strong> at Zosano.<br />

In October 2011, Zosano Pharma<br />

announced a long-term strategic<br />

collaboration with Asahi Kasei Pharma<br />

Corporation (AKP) for the development,<br />

commercialization, and supply of a weekly<br />

patch formulation of TeriboneTM (human<br />

PTH 1-34). As part of this Asian licensing<br />

agreement, AKP has paid Zosano $7.5<br />

million in upfront consideration. In addition,<br />

AKP will pay more than $25 million in<br />

milestone payments related to development,<br />

regulatory, and product launch. Zosano will<br />

receive revenue-based royalties based on<br />

sales of the ZP patch formulation of<br />

Teribone in the Asian territories, as well as<br />

reimbursement for all development and<br />

manufacturing costs and commercialization.<br />

AKP is committing significant additional<br />

financial and technical resources toward the<br />

development of the patch for the treatment<br />

of osteoporosis in the Asian territories, and<br />

has already commenced clinical trials, which<br />

are now entering the pivotal Phase III stage.<br />

Zosano retains US/Europe licensing<br />

rights of ZP patch development of PTH 1-<br />

34 and expects to further capitalize on the<br />

momentum gained by its recent<br />

announcement on this and other products.<br />

In short, there will be more choices<br />

available to the patient than ever before<br />

regarding therapeutics and the delivery<br />

methods all focused on greater efficacy and<br />

improved convenience/compliance, says Mr.<br />

Ripple.<br />

“We predict a game-changing moment<br />

once the first effective needle free, pain-free<br />

drug delivery product enters various markets<br />

and the expectation of convenient, needlefree<br />

drug delivery is established,” he<br />

concludes.<br />

THE FUTURE OF<br />

TRANSDERMAL, TOPICAL<br />

& SUBCUTANEOUS<br />

DRUG DELIVERY<br />

Looking forward, the pros predict that<br />

future growth rate for transdermal, topical,<br />

and subcutaneous products will be slow to<br />

moderate. The primary reason for no real<br />

“revolutionary” products or technologies in<br />

the near-term future is the fact that most<br />

pharma and biotech companies continue to<br />

be very conservative when it comes to earlystage<br />

development and associated risk. Add<br />

to that the FDA’s conservative stance toward<br />

adopting and approving new drugs or new<br />

delivery systems/technologies, and you have<br />

another significant growth limiting factor.<br />

Throw in the financial cliff facing the US by<br />

year’s end and the future of nationalized<br />

healthcare, and the picture becomes rather<br />

murky. u<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

57


Gail Schulze<br />

CEO & Executive Chair<br />

Thomas of the Board Harlan<br />

CEO<br />

Zosano<br />

Caisson Biotech<br />

“Caisson’s HepTune involves<br />

the process of conjugating<br />

this naturally occurring<br />

sugar molecule, heparosan,<br />

to drugs. The size of the<br />

heparosan and the<br />

conjugation method for<br />

coupling can vary depending<br />

on the drug cargo, the<br />

client’s/partner’s preference,<br />

and achievement of optimal<br />

performance. Caisson works<br />

to meet the partner’s<br />

clinical performance needs<br />

for each drug conjugate.”<br />

CAISSON BIOTECH: <strong>IN</strong>NOVATION <strong>IN</strong><br />

DRUG DELIVERY US<strong>IN</strong>G A NATURALLY<br />

OCCURR<strong>IN</strong>G SUGAR MOLECULE<br />

Motivated by increasingly reported adverse events related to PEGylated drugs and<br />

based on the pioneering glycobiology research of Dr. Paul DeAngelis of the<br />

University of Oklahoma Health Science Center (OUHSC), Caisson Biotech, L.L.C.<br />

has developed and patented a drug delivery technology utilizing naturally occurring sugar<br />

polymers to more safely and effectively deliver drugs. Customizable to any currently PEGylated<br />

drug or to any other molecule requiring this type of stealth modification, HepTuneTM , Caisson’s<br />

drug delivery system, can be used in the treatment of a number of diseases. Caisson’s patented<br />

delivery system harnesses the naturally occurring sugar molecule, heparosan, as its delivery<br />

vehicle. By utilizing proprietary methods to manufacture heparosan and its derivatives, Caisson<br />

offers its HepTune as a safer delivery vehicle in general and specifically, as an alternative to<br />

PEG (poly[ethylene glycol]). HepTune has many bio-superior attributes over PEGylation,<br />

including greater compatibility, lack of accumulation in tissues, extremely desirable PK and<br />

safety profiles, and no known toxic effects. Additionally, conjugation of Caisson’s heparosanbased<br />

reagent affords new and novel intellectual property either for new molecules or to extend<br />

the patent life of already-marketed drugs, and also recaptures the percentage of the market due<br />

to patients that have developed PEG sensitivity excluding them from current PEGylated<br />

formulations. <strong>Drug</strong> Developemnt & <strong>Delivery</strong> recently spoke with Thomas Harlan, CEO of<br />

Caisson, to discuss how his company is improving the quality and delivery of numerous<br />

medications, making life easier for patients, and offering new ways for companies to enhance<br />

their drug pipeline.<br />

Q: Can you please provide our<br />

readers some background on<br />

Caisson Biotech?<br />

A: Caisson is a portfolio company of Emergent<br />

Technologies, Inc. (ETI), a leading technology<br />

access and innovation management company<br />

(www.etibio.com). ETI applies rigorous selection<br />

criteria to assess and evaluate technologies<br />

emanating from thought-leading scientists in<br />

universities and federal research labs. Emergent<br />

then builds commercially viable businesses based<br />

on those platform technologies. ETI has innovated<br />

the start-up company management model, allowing<br />

for effective initial technology transfer, rapid rampup<br />

or scale-back of business (depending on the<br />

maturity of the technology), and technical resources<br />

in response to market needs, aggressive intellectual<br />

property (IP) protection, and IP scope expansion<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

59


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

60<br />

and G&A expense cost-sharing. ETI scouted<br />

the glycobiology research of Dr. DeAngelis,<br />

and with him (licensing the technology from<br />

OUHSC) built a family of companies to<br />

produce and explore applications for three<br />

commercially important glycosaminoglycan<br />

(GAG) polymers: hyaluronic acid (HA),<br />

heparosan, and chondroitin. Collectively,<br />

these companies (Hyalose, L.L.C.,<br />

Heparinex, L.L.C., and Choncept, L.L.C.)<br />

have partnered with pharmaceutical and<br />

biotechnologyy companies to evaluate GAGs<br />

for application areas ranging across<br />

rheumatology, ophthalmology, tissue<br />

engineering, dermal fillers and reconstructive<br />

surgery, drug delivery, biomaterials, medical<br />

device coating, and anti-adhesion films.<br />

Caisson was formed in 2009 as a wholly<br />

owned subsidiary of Heparinex when Dr.<br />

DeAngelis’ research on heparosan, in<br />

particular, began to show bio-superior<br />

properties over PEG for drug delivery.<br />

Q: What is it about heparosan<br />

that makes it Caisson’s drug<br />

delivery vehicle of choice?<br />

A: Heparosan is structurally related to<br />

heparin, one of the most widely used drugs in<br />

the Pharmacopeia. Heparosan was predicted<br />

to be biocompatible in the human body as it<br />

is a natural precursor in the heparin<br />

biosynthetic pathway and also because of the<br />

stretches of heparosan that exist in human<br />

heparan sulfate chains. In addition, certain<br />

pathogenic bacteria even use a heparosan<br />

coating to evade the immune system during<br />

infection. Caisson’s HepTune involves the<br />

process of conjugating this naturally<br />

occurring sugar molecule, heparosan, to<br />

drugs. The size of the heparosan and the<br />

conjugation method for coupling can vary<br />

depending on the drug cargo, the<br />

client’s/partner’s preference, and achievement<br />

of optimal performance. Caisson works to<br />

meet the partner’s clinical performance needs<br />

for each drug conjugate.<br />

Q: Many companies focus on<br />

drug delivery technologies.<br />

What makes Caisson’s HepTune<br />

unique, and how does is it differ<br />

from PEGylation?<br />

A: A significant challenge in the<br />

pharmaceutical industry exists in which drugs<br />

are excreted too quickly from the body. This<br />

can cause patients to endure an increased<br />

number of treatment injections (requiring<br />

repeated, either multiple injections in a single<br />

day or daily injections over an extended<br />

period of time) and generate strong<br />

immunogenic responses. Currently, PEG is<br />

the most widely used drug delivery agent for<br />

protein-based drugs to overcome these<br />

problems, but suffers from liabilities,<br />

including detrimental accumulation in organs<br />

and antigenicity of its own. Furthermore,<br />

higher doses and/or lifetime treatments using<br />

PEG could amplify these problems because<br />

the liver detoxification system creates a<br />

variety of reactive PEG metabolites that are<br />

cytotoxic. There is a rising occurrence of<br />

PEG immunogenicity. In 1984, anti-PEG<br />

antibodies were detected in 0.2% of naïve<br />

patients sampled, but as of 2001, stunningly<br />

22% to 25% of healthy blood donor samples<br />

(n = 350) had anti-PEG antibodies. 1 It is<br />

thought this increase of anti-PEG antibodies<br />

in the general population is due to the<br />

increasing use of PEG in consumer products,<br />

such as toothpaste, laxatives, vitamin pills,<br />

and many other products commonly used on<br />

a daily basis. Indeed, some childhood<br />

leukemia patients no longer respond to their<br />

PEGylated asparaginase (Oncaspar ® )<br />

medication due to anti-PEG antibody levels. 2<br />

In contrast, the natural heparosan polymers of<br />

Caisson’s HepTune have the superior<br />

synergistic combination of biocompatibility,<br />

lack of immunogenicity, and long half-life in<br />

the bloodstream. Caisson’s heparosanreagents<br />

are degraded into normal sugars and<br />

even recycled into other molecules the body<br />

uses and thus possess substantially lower<br />

toxicity.<br />

Furthermore, the quality control of PEG<br />

polymer synthesis (ie, the drug delivery<br />

vehicle) with respect to molecular weight<br />

distribution is less than optimal. The length of<br />

the linear polymer is not uniform and<br />

typically a preparation of linear PEG<br />

polymers greater than 10 kDa, a portion<br />

(~3%) of chains might have branching. This<br />

unplanned branching can yield a series of<br />

cross-linked complexes containing multiple<br />

cargo molecules because every branch has a<br />

reactive group for coupling to the cargo. On<br />

the other hand, linear heparosan reagents are<br />

substantially uniform in length, even for<br />

masses up to 800 kDa. In addition, due to an<br />

innovative synthesis method involving<br />

enzymatic polymerization, Caisson’s<br />

heparosan drug delivery vehicles cannot be<br />

branched and can never have more than a<br />

single reactive group. The attachment of the<br />

heparosan vehicle to drug cargo, has many<br />

other superior attributes over PEGylation,<br />

including ease of generating a larger size<br />

range of polymers, higher water solubility,<br />

greater biocompatibility of degradation<br />

products, lack of accumulation in tissues, and<br />

new intellectual property. In a 2008 Current<br />

Opinions in <strong>Drug</strong> Discovery & <strong>Development</strong><br />

article, it was predicted future drugs will use<br />

higher molecular weight PEGs and/or be<br />

given at higher doses for long periods.


Caisson predicts heparosan will be the<br />

preferable therapeutic vehicle based on<br />

supporting preliminary study results and due<br />

to PEG’s intrinsic limitations and emerging<br />

immunogenicity.<br />

Q: What impact do you see<br />

Caisson Biotech’s HepTune<br />

technology having in the<br />

market?<br />

A: Caisson Biotech is well positioned to<br />

have a significant impact in the market in a<br />

number of ways. Caisson has entered into<br />

the market with a naturally occurring sugar<br />

polymer drug delivery vehicle, HepTune,<br />

with many superior performance benefits<br />

when compared to current existing<br />

competitive drug delivery systems.<br />

Supporting this technology is a very strong<br />

patent portfolio. Caisson owns or has rights<br />

to 13 patents, 4 of which are issued.<br />

Caisson’s aforementioned patents are a<br />

subset of patents within Dr. DeAngelis’ total<br />

patent portfolio for carbohydrate production<br />

consisting of over 200 patents and patent<br />

applications.<br />

The Caisson patent claims consist of<br />

multiple and distinct heparosan production<br />

methodologies, the use of heparosan as a<br />

biomaterial and also the use of heparosan<br />

conjugated to therapeutics. PEG, the base<br />

material for the PEGylation platform, is a<br />

publicly available material. Unlike the<br />

innovators of the PEGylation technology,<br />

Caisson retains rights to the composition-ofmatter<br />

and methods-of-manufacture of the<br />

heparosan conjugation reagent. This<br />

includes the production of monodisperse<br />

heparosan materials. The ability to have<br />

claim coverage related to the production of<br />

the base material (ie, heparosan) provides<br />

added protection and exclusivity around the<br />

ultimate conjugated materials and<br />

therapeutic products. In addition, Caisson<br />

benefits from the rights to the control of<br />

production and supply of the heparosan<br />

material.<br />

With the many advantages outlined,<br />

Caisson believes it is well positioned to<br />

make a significant impact and contribution<br />

to the current drug delivery market, greatly<br />

benefiting pharmaceutical companies and<br />

ultimately patients.<br />

Q: What makes Caisson<br />

Biotech an ideal partner?<br />

A: Caisson Biotech has a commercially<br />

proven drug delivery technology, a highly<br />

motivated and committed staff, and a track<br />

record of meeting milestones specified in<br />

contracts, both commercial and grant<br />

sourced. Caisson’s employees have a<br />

combined 66 years of experience in the<br />

Glycobiology field ranging from the<br />

discovery and manipulation of enzymes<br />

responsible for synthesis of GAG polymers<br />

(heparosan, chondroitin, and hyaluronic<br />

acid) to production and validation of<br />

heparosan for use in drug delivery. Caisson’s<br />

founding scientist, Dr. Paul DeAngelis, is<br />

highly recognized in the Glycobiology field<br />

for various contributions, both scientifically<br />

and for his commercial success, and is still<br />

actively engaged with Caisson on a daily<br />

basis. Companies interested in evaluating<br />

HepTune will find the management easy to<br />

work with, the cost of evaluation very<br />

affordable, and the process whereby<br />

conjugates are specified and procured for<br />

evaluation and testing easy to comprehend<br />

and comply with. The corporate<br />

management has developed a simple, timely,<br />

and effective engagement protocol for<br />

companies interested in evaluating the<br />

technology.<br />

Q: What can we expect to see<br />

from Caisson in the market?<br />

A: In May, 2012 Caisson announced Novo<br />

Nordisk as its first commercialization<br />

partner. The recently executed<br />

<strong>Development</strong>al License Agreement focuses<br />

on development of a number of heparosanconjugated<br />

drugs. One or more of these<br />

drugs are projected to enter into clinical<br />

trials between 2013 and 2014.<br />

Caisson is also developing an internal<br />

drug pipeline with several products currently<br />

progressing through preclinical trials. The<br />

company is gearing up for commercial-scale<br />

production to handle multiple new clients<br />

and expects to see active sales and<br />

marketing efforts by its partners in the US<br />

and Europe in the near future. Caisson looks<br />

forward to the rapid advancement of its<br />

clinical pipeline and the opportunity to work<br />

with additional industry partners to bring<br />

novel therapeutics to patients in need.<br />

References<br />

1. Garratty G. Progress in modulating the<br />

RBC membrane to produce transfusable<br />

universal/stealth donor RBCs. Trans Med<br />

Rev. 2004;18:245-256.<br />

2. Armstrong JK, et al. Antibody against<br />

poly(ethylene glycol) adversely affects<br />

PEG-asparaginase therapy in acute<br />

lymphoblastic leukemia patients. Cancer.<br />

110:103-111.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

61


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

62<br />

TRANSDERMAL SYSTEMS & COMPONENTS<br />

Companies worldwide<br />

look to 3M <strong>Drug</strong><br />

<strong>Delivery</strong> Systems for<br />

ingenious transdermal<br />

systems and<br />

components as well as<br />

manufacturing solutions<br />

to help accelerate<br />

development and<br />

enable success. 3M<br />

can fulfill all your<br />

transdermal delivery<br />

needs, including new microneedle technology. 3M’s microneedle platform,<br />

Microstructured Transdermal Systems (MTS) leverage core 3M<br />

competencies to expand the range of transdermally deliverable drugs to<br />

include proteins, peptides, and vaccines. MTS can also deliver significant<br />

benefits over injection, including improved delivery efficiency and faster<br />

absorption of some drugs and vaccines as well as the potential to induce<br />

similar maximum drug concentrations at a significantly lower dose. Please<br />

view our latest poster presentation, The Transdermal <strong>Delivery</strong> of Human<br />

Growth Hormone, at www.3M.com/tddpublications. For more information,<br />

call 3M DDS at (800) 643-8086 (US) or visit www.3M.com/MTS.<br />

CONTRACT TEST<strong>IN</strong>G LABORATORY<br />

Advantar Labs is a top-tier cGMP and GLP Contract Testing Laboratory<br />

catering to the pharmaceutical, biopharmaceutical, and medical devices<br />

industries. We offer an exceptional portfolio of scientific services,<br />

including Method <strong>Development</strong> and Validation, Method Remediation,<br />

Preformulation and Formulation <strong>Development</strong>, Litigation Support, as well<br />

as Extractables and Leachables. We also provide customized smallscale<br />

Clinical Trial Packaging and ICH Stability Storage services. Our<br />

elite team of scientists enables you to increase your clinical success by<br />

optimizing your CMC strategy and reducing the potential for mistakes<br />

through our highly efficient lab operations, study protocols, and data<br />

management systems. For more information, contact Advantar Labs at<br />

(858) 228-7788 or visit www.advantarlabs.com.<br />

DRUG DEVELOPMENT SERVICES<br />

AAIPharma Services Corp. is a leading provider of services that<br />

encompass the entire pharmaceutical drug development process from<br />

early development through commercialization. The company, which has<br />

the benefit of more than 30 years of experience, specializes in analytical<br />

chemistry, formulation development, clinical packaging, oral drug delivery,<br />

and contract manufacturing. Headquartered in Wilmington, NC, AAIPharma<br />

Services serves more than 300 large pharmaceutical and biotechnology<br />

companies. Our clients say we are experienced, responsive, and<br />

dependable in: starting rapidly, meeting timelines, seeing obstacles before<br />

they occur, and responding quickly when things change. Let us move your<br />

project forward reliably. For more information, contact AAIPharma Services<br />

Corp. at (800) 575-4224, email at services@aaipharma.com, or visit<br />

www.aaipharma.com.<br />

LICENS<strong>IN</strong>G & CAPABILITIES<br />

Aveva has a number of products for license from its development<br />

pipeline along with a full complement of R&D capabilities to produce<br />

transdermal drug delivery systems that fortify pipelines and maximize<br />

product life cycles. Aveva <strong>Drug</strong> <strong>Delivery</strong> Systems is one of the world’s<br />

largest manufacturers of, and a pioneer in, transdermal drug delivery<br />

systems with a rich history of providing pharmaceutical partners with<br />

fully integrated, controlled-release transdermal products that fulfill<br />

unmet market needs. Products for licensing include Sufentanil,<br />

Fentanyl, Clonidine, and Nicotine. For more information, contact<br />

Robert Bloder, VP of Business <strong>Development</strong>, at (954) 624-1374 or<br />

visit www.avevadds.com.


SOLUBILIZER COMPENDIUM<br />

BASF’s new solubilizer<br />

compendium is a must-read for<br />

anyone working with APIs that<br />

exhibit poor solubility and<br />

bioavailability. It leverages BASF’s<br />

vast expertise in solubilization and<br />

bioavailabilty enhancement, and is<br />

the result of many years of<br />

research. The publication provides<br />

a valuable overview of all relevant<br />

BASF excipients (Kolliphor TM<br />

grades, Soluplus ® , and selected<br />

Kollidon ® grades), and offers<br />

helpful advice on creating solid<br />

solutions and dispersions. What’s<br />

more, it includes a chapter<br />

dedicated to high-throughput<br />

screening as a means of selecting<br />

the right excipient or combination of excipients for a poorly soluble drug.<br />

Visit www.innovate-excipients.basf.com to download the compendium as<br />

a PDF, or to request a free hard copy. BASF’s solubility enhancement<br />

experts are happy to answer all your questions. Just send an e-mail to<br />

pharma-ingredients@basf.com.<br />

PHARMACEUTICAL SOLUTIONS<br />

Catalent Pharma Solutions is a world leader in patented drug delivery<br />

technologies. For more than 70 years, we have developed and<br />

manufactured advanced drug delivery systems and partnered with<br />

nearly every major global pharmaceutical company. We continually work<br />

to advance the science of drug delivery and enhance the therapeutic<br />

and market performance of our customers’ drugs. Our advanced drug<br />

delivery technologies bring new options to resolve the technical<br />

challenges development scientists face every day. These patented<br />

technologies can improve the odds of successful formulation by<br />

enhancing bioavailability, optimizing the rate of release, and targeting<br />

the site of absorption. Our technologies include softgel and Vegicaps ®<br />

Soft capsules; Zydis ® fast-dissolve dosage form; modified-release<br />

technologies; and a range of inhaled technologies, including MDIs, DPIs,<br />

nasal sprays, and solutions/suspensions for inhalation, nebulizers, and<br />

liquid inhalers. For more information, contact Catalent Pharma Solutions<br />

at (866) 720-3148 or visit www.catalent.com.<br />

DEVELOPMENT & MANUFACTUR<strong>IN</strong>G<br />

Bend Research is a leading independent scientific development and<br />

manufacturing company with more than 35 years of experience in the<br />

development of pharmaceutical delivery systems. To achieve its mission of<br />

improving health through the advancement of its clients’ best new<br />

medicines, the company uses a multidisciplinary problem-solving<br />

approach grounded in science and engineering fundamentals to address<br />

the most difficult challenges. As a leader in novel drug delivery<br />

technologies and formulations, Bend Research provides expertise in<br />

solubilization technologies, such as spray-dried dispersions and hot-melt<br />

extrusion formulations, as well as biotherapeutic, controlled-release,<br />

inhalation, and nanoparticle technologies. Capabilities include formulation<br />

science, process development and engineering, dosage-form<br />

development, cGMP manufacturing, and analytical services for the<br />

advancement of drug candidates from early discovery to<br />

commercialization. Bend Research also pioneers, advances, and<br />

commercializes new technologies. For more information, contact Phoenix<br />

Ivers of Bend Research at (800) 706-8655 or<br />

Phoenix.Ivers@BendResearch.com or visit www.BendResearch.com.<br />

COMB<strong>IN</strong>ATION CAPSULE TECHNOLOGY<br />

InnerCap offers an advanced<br />

patented multi-phased, multicompartmentalizedcapsularbased<br />

delivery system. The<br />

system can be used to enhance<br />

the value and benefits of<br />

pharmaceutical and<br />

biopharmaceutical products.<br />

Utilizing two-piece hard shell<br />

capsules, the technology offers<br />

the industry solutions to<br />

problems affecting<br />

pharmaceutical companies,<br />

patients, and healthcare<br />

providers. The delivery system<br />

will be licensed to enhance<br />

pharmaceutical and<br />

biopharmaceutical products. It is a very effective way to deliver<br />

multiple active chemical compounds in different physical phases with<br />

controlled-release profiles. The delivery system provides the<br />

pharmaceutical and biopharmaceutical industries with beneficial<br />

solutions to the industry’s highly publicized need to repackage and<br />

reformulate existing patented blockbuster drugs with expiring patents<br />

over the next 5 years. For more information, contact InnerCap<br />

Technologies, Inc., at (813) 837-0796 or visit www.innercap.com.<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

63


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

64<br />

MARKET<strong>IN</strong>G & COMMUNICATIONS<br />

Get Noticed. Get<br />

Funded. Grow<br />

Faster. When you<br />

need to connect<br />

with investors,<br />

business<br />

partners, and<br />

regulatory<br />

agencies,<br />

LifeSciencePR<br />

can make that<br />

happen. Our<br />

integrated communication strategies and well-established industry<br />

contacts will help your life science company achieve its short- and longterm<br />

corporate objectives. We work seamlessly with your senior<br />

management team to develop the most effective communication initiatives<br />

to reach your prospective investors and partners. Our experienced staff<br />

knows what it takes to break through with your breakthroughs, powering<br />

your engine in your continued drive toward your success. LifeSciencePR<br />

will get you there smarter, faster, and easier than any other marketing and<br />

communications firm in the industry. For more information, contact<br />

LifeSciencePR at (800) 724-2372 or visit www.LifeSciencePR.net.<br />

TUNABLE HALF-LIFE TECHNOLOGY<br />

Novozymes’ tunable<br />

half-life technology<br />

has been developed<br />

to flexibly tune the<br />

pharmacokinetic<br />

profile of target<br />

proteins and<br />

peptides. The halflife<br />

of albumin can<br />

be tuned up or<br />

down to meet the half-life needs of the customer’s drug and application.<br />

The technology is based on the interaction between albumin and the<br />

neonatal Fc receptor (FcRn) to extend circulatory half-life. Building on<br />

Novozymes’ knowledge of albumin and its native receptor, albumin can<br />

be modified to increase receptor binding and, consequently, deliver an<br />

increase in the pharmacokinetics of fused or conjugated therapeutics.<br />

The Flex technology offers the potential to increase half-life according to<br />

specific medical needs. This allows delivery of drugs with extended<br />

circulatory time, therefore reducing dosing regimes and increasing<br />

patient compliance. For more information on Novozymes’ Flex<br />

technology, visit www.biopharma.novozymes.com.<br />

PLATFORM TECHNOLOGY<br />

Ligand is a biopharmaceutical company that develops and acquires<br />

technology and royalty revenue generating assets that are coupled to a<br />

lean cost structure. Ligand’s Captisol ® platform technology is a patent<br />

protected, chemically modified cyclodextrin with a structure designed to<br />

optimize the solubility and stability of drugs. Captisol ® has enabled five<br />

FDA-approved products, including Pfizer’s VFEND ® IV and Baxter’s<br />

Nexterone ® . For licensing opportunities, call Captisol Services at (877)<br />

575-5593 or visit www.captisol.com.<br />

SILICONE MATERIALS<br />

When it comes to drug delivery, NuSil provides numerous solutions<br />

that fit a variety of device needs. While most silicone products are<br />

customized for individual delivery systems, all are developed with<br />

FDA regulatory concerns in mind. In addition to its role as a supplier,<br />

NuSil offers research and development capabilities for those looking<br />

for proprietary, custom formulations. Regardless of batch size, NuSil<br />

delivers quality, high-performance silicone materials based on your<br />

unique property requirements, as well as provides precise, custom<br />

formulations. NuSil offers an even wider range of silicone material<br />

and compound options for transdermal, transmucosal, implanted<br />

intrathecal, and external delivery devices, as well as ingestible<br />

materials. For more information, contact NuSil Technology at (805)<br />

684-8780 or visit www.nusil.com.


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

66<br />

KNOWLEDGE MANAGEMENT<br />

PharmaCircle is<br />

an innovative<br />

knowledge<br />

management<br />

company<br />

specializing in<br />

the drug<br />

delivery,<br />

pharmaceutical,<br />

and<br />

biotechnology<br />

fields, with a current client base ranging from start-up life science<br />

companies to world leaders in Big Pharma. Clients choose PharmaCircle’s<br />

services and content for its comprehensive technical (pipeline, products,<br />

molecule, and technology) and business (deals, acquisitions, royalty,<br />

licensing, drug revenues, market information, etc) related information and<br />

analysis, which are ideal for all segments of small and large companies.<br />

PharmaCircle helps facilitate product life cycle management (LCM),<br />

partnering, licensing, and competitive intelligence efforts as well as<br />

supplements internal efforts and costs at a fraction of the cost if<br />

performed internally. For more information, contact PharmaCircle at (920)<br />

850-3056 or visit www.pharmacircle.com.<br />

CONTRACT DIAGNOSTICS<br />

ResearchDx is the leading Contract Diagnostics Organization (CDO) for the<br />

biopharmaceutical and diagnostic industries. As a CDO, ResearchDx<br />

integrates in vitro diagnostics (IVD) non-clinical and clinical research,<br />

CAP/CLIA laboratory assay discovery, development and testing, GMP<br />

manufacturing of test kit components, and end-to-end consulting to<br />

achieve the fastest path to market for your IVD/companion diagnostic.<br />

With experts managing the entire development process from initial assay<br />

concept and discovery through clinical research to regulatory approval,<br />

ResearchDx employs a proactive project management approach with the<br />

availability of resources to ensure appropriate attention to timelines every<br />

step of the way. By offering the complete range of CDO services without<br />

owning a proprietary testing platform, ResearchDx ensures its clients’<br />

diagnostics are developed with the methodology and platform that best<br />

optimize its characteristics and commercialization strategy. For more<br />

information, contact ResearchDx at (866) 225-9195 or visit<br />

www.researchdx.com.<br />

DEVELOPMENT SERVICES DEVELOPMENT SERVICES<br />

UPM Pharmaceuticals ® is an independent provider of contract<br />

formulation development, analytical services, and cGMP manufacturing.<br />

We continue a legacy of intellectual distinction and uncompromising<br />

performance with every new project. The talent and experience of our<br />

team, our dedication to science-based formulation design, and our<br />

commitment to communication and timeliness enables us to offer the<br />

highest level of customized drug development services. Our 40,000-sqft<br />

main facility in Baltimore features cGMP pharmaceutical<br />

manufacturing and packaging suites as well as analytical and R&D<br />

laboratories staffed by industry veterans. Whatever form your product<br />

takes, we ensure rigorous and technically sound product<br />

characterization, methods development, and QC release. Our clients<br />

enjoy service that is highly responsive and fast with total quality<br />

management characteristic of a customer-focused business. For more<br />

information, contact UPM Pharmaceuticals at 410-843-3738 or visit<br />

www.upm-inc.com.<br />

Xcelience is a premier provider of formulation development and<br />

manufacturing solutions with a solid reputation for accelerating early<br />

phase small molecule development. Our outstanding quality record,<br />

significant drug development expertise, willingness to customize, and<br />

disciplined project management enable us to deliver real advantages<br />

to clients needing to speed potential drugs to clinical trials. Since<br />

1997, Xcelience has been renowned for reliably expediting drug<br />

development. Our formulation development scientists have<br />

considerable experience overcoming challenges associated with<br />

physical and chemical properties of drug substance, or limited<br />

quantities of API, in a manner that results in compounds with<br />

improved solubility and bioavailability. Partnering with a specialist like<br />

Xcelience for early phase development can significantly reduce<br />

product risk and accelerate development timelines. For more<br />

information, contact Xcelience at (608) 643-4444 or visit<br />

www.xcelience.com.


<strong>Drug</strong><br />

<strong>Development</strong><br />

Deuterium Modification as a New Branch of<br />

Medicinal Chemistry to Develop Novel, Highly<br />

Differentiated <strong>Drug</strong>s<br />

By: Philip Graham, PhD; Julie Liu, PhD; and David Turnquist, MBA; Concert Pharmaceuticals, Inc.<br />

Introduction<br />

Concert Pharmaceuticals, a clinical<br />

stage biopharmaceutical company, uses<br />

deuterium to create and develop highly<br />

differentiated new medicines. This is a<br />

significant departure from the traditional use<br />

of deuterium modification as an analytical<br />

probe for metabolic studies. Concert uses its<br />

DCE PlatformTM (Deuterium Chemical<br />

Entity) to develop new drugs that are<br />

designed to have first-in-class or best-inclass<br />

efficacy and/or safety. Deuterium<br />

modification provides a novel opportunity to<br />

develop new drugs by building upon existing<br />

scientific or clinical experience, potentially<br />

significantly reducing time, risk, and<br />

expense.<br />

Concert’s DCE Platform is based on the<br />

premise of the deuterium isotope effect<br />

wherein selective replacement of hydrogen<br />

atoms with deuterium creates more stable<br />

chemical bonds with carbon atoms. This<br />

modification can sometimes improve drug<br />

metabolism, increase half-life, or enhance<br />

bioavailability and exposure in a significant<br />

fashion. Deuterium is a safe, non-radioactive,<br />

naturally abundant isotope of hydrogen. The<br />

body of the average human adult already<br />

contains about 2 g of deuterium. Due to its<br />

similarity to hydrogen, deuterium<br />

modification usually has negligible effect on<br />

the intrinsic activity of a drug at its<br />

biological target. Concert’s deuterium<br />

modification drug development approach is<br />

thus a powerful tool to improve the ADME<br />

(absorption, distribution, metabolism, and<br />

elimination) properties of a drug while<br />

maintaining its intrinsic biological activity.<br />

As many drugs under development or on the<br />

market suffer from sub-optimal<br />

pharmacokinetic and metabolic profiles,<br />

deuterium modification offers great promise<br />

to improve the profiles of these drugs and<br />

open opportunities for new uses.<br />

Concert has advanced a number of<br />

deuterium-containing novel compounds<br />

designed to have unique therapeutic<br />

properties. The company has made<br />

preclinical and clinical progress with<br />

proprietary drug compounds in diverse<br />

therapeutic areas, including potential<br />

treatments for diabetic nephropathy, hot<br />

flashes, spasticity, neuropathic pain, and<br />

multiple myeloma.<br />

CTP-499 for Diabetic<br />

Nephropathy<br />

Concert’s lead drug candidate is CTP-<br />

499, a novel potential treatment for diabetic<br />

nephropathy in type 2 diabetics. Diabetic<br />

nephropathy is the leading cause of endstage<br />

renal disease, or kidney failure, and is<br />

expected to grow significantly as the<br />

incidence of type 2 diabetes increases<br />

rapidly. Despite the availability of blood<br />

pressure lowering agents, such as angiotensin<br />

II receptor blockers (ARBs) and angiotensinconverting<br />

enzyme inhibitors (ACEIs), many<br />

patients continue to experience a decline in<br />

renal function and progress to kidney failure.<br />

As a result, there is a critical need for new<br />

drugs with untapped mechanisms that can<br />

further delay or prevent the decline of kidney<br />

function and eventual need for dialysis.<br />

CTP-499 is an analog of 1-((S)-5hydroxyhexyl)-3,7-dimethylxanthine<br />

(HDX),<br />

an active metabolite of Trental ®<br />

(pentoxifylline), that Concert created by<br />

SPECIALTY PHARMA JULY/AUGUST 2012 Vol 12 No 6<br />

67


eplacing several key hydrogen atoms with<br />

deuterium. Trental, which is significantly<br />

metabolized to HDX, is approved in the US<br />

for the treatment of intermittent claudication<br />

and has been reported in preliminary studies<br />

to have beneficial effects on urinary albumin<br />

excretion and renal function decline. Based<br />

on the pharmacology and degree of exposure,<br />

it appears these renoprotective effects may be<br />

due in large part to HDX. CTP-499 and HDX<br />

have similar physical, chemical, and<br />

pharmacological properties; however, Phase I<br />

clinical data with CTP-499 shows that<br />

incorporation of deuterium leads to increased<br />

exposure to the species responsible for the<br />

renoprotective effects.<br />

To date, four Phase I studies have been<br />

completed with CTP-499. One study showed<br />

that controlled-release (CR) formulations of<br />

CTP-499 form the same metabolites as<br />

Trental; however, exposure to species<br />

possessing beneficial anti-inflammatory, anti-<br />

fibrotic, and immunomodulatory effects was<br />

increased with CTP-499 relative to Trental<br />

administration. The overall pharmacokinetic<br />

profile suggested CR CTP-499 may be<br />

suitable for once-daily dosing. A second<br />

study, which tested single ascending doses of<br />

CR CTP-499, showed that doses up to and<br />

including 1800 mg were well tolerated. Based<br />

on these results, CTP-499 shows unique<br />

potential as a treatment for diabetic<br />

nephropathy, along with reduced development<br />

risk given the abundant safety experience<br />

with Trental.<br />

In early 2012, Concert initiated a<br />

6-month Phase II efficacy study of CTP-499<br />

in diabetic patients with mild-to-moderate<br />

renal impairment who are at particular risk of<br />

disease progression due to macroalbuminuria<br />

(excessive amounts of albumin in their urine).<br />

The primary goal of the study is to<br />

investigate the effect of CTP-499 on a<br />

measure of disease progression, urinary<br />

albumin excretion, with secondary goals<br />

related to safety/tolerability and effects on<br />

renal function and various biomarkers.<br />

The impact of deuterium on cost of<br />

goods varies depending on the drug. Concert<br />

invested in process development research that<br />

resulted in an elegant, highly efficient<br />

manufacturing process for CTP-499 active<br />

pharmaceutical ingredient (API). The<br />

synthesis has been designed to use the most<br />

cost-effective deuterated starting material and<br />

produces API in > 70% yield. Because CTP-<br />

499 is a single enantiomer, the desired<br />

enantiomer is directly isolated using an<br />

asymmetric synthesis with high<br />

enantioselectivity and little loss of deuterium.<br />

Finally, Concert creatively implemented<br />

recycling and recovery techniques through<br />

which > 90% of the deuterium used is either<br />

incorporated into the API or recovered for<br />

future use. CTP-499 is manufactured on a<br />

scale of hundreds of kilograms and is<br />

expected to have a cost of goods that is<br />

competitive with other commercial APIs.<br />

CTP-347: Deuterium<br />

Modification of<br />

Paroxetine<br />

Concert’s first clinical candidate was<br />

CTP-347, a selectively deuterated analogue of<br />

paroxetine. CTP-347 was designed to<br />

eliminate the irreversible inhibition of a key<br />

metabolizing enzyme (CYP2D6) caused by a<br />

highly reactive paroxetine metabolite that<br />

covalently binds to the enzyme. In certain<br />

patients potentially benefiting from therapy<br />

with paroxetine who also receive endocrine<br />

disrupting agents, such as post-menopausal<br />

women and cancer patients, paroxetine use<br />

can be complicated or contraindicated as it<br />

causes extensive drug-drug interactions<br />

(DDIs) with other medications. In vitro<br />

metabolism experiments with CTP-347<br />

demonstrated little to no CYP2D6<br />

inactivation, as a result of deuterium-based<br />

metabolic shunting, which essentially<br />

prevented the formation of the reactive<br />

metabolite. Other studies showed the intrinsic<br />

pharmacology of paroxetine was unchanged<br />

after deuterium incorporation. A 96-subject<br />

single and multiple ascending dose clinical<br />

study with CTP-347 provided further<br />

evidence, consistent with the in vitro data,<br />

that irreversible inactivation of the CYP2D6<br />

enzyme did not occur with CTP-347. This<br />

clinical study with CTP-347 represents the<br />

first clinical demonstration that deuteration<br />

can be utilized to avoid the formation of<br />

undesired metabolites in humans.<br />

CTP-354: A Deuterium-<br />

Modified Subtype-<br />

Selective GABAA Modulator<br />

Concert has created deuterated subtypeselective<br />

GABAA modulators that represent a<br />

promising therapeutic modality for the<br />

treatment of spasticity and chronic pain, with<br />

the potential for improvement over existing<br />

first-line therapies. A subtype-selective<br />

GABAA modulator has the potential to<br />

address a major unmet need by retaining<br />

desirable therapeutic aspects of the<br />

benzodiazepines - anxiolytic and spasmolytic<br />

activity - while minimizing the sedative and<br />

tolerance effects.<br />

Concert’s GABAA modulator program is<br />

based upon agents that have been profiled in<br />

multiple preclinical efficacy models,<br />

representing an opportunity to leverage<br />

extensive therapeutic data to advance a<br />

clinical candidate. Concert’s GABAA modulator lead compound, CTP-354, is a<br />

deuterated version of a preclinical agent<br />

discovered at Merck. The Merck compound,<br />

L-838417, was targeted by Concert for<br />

deuterium substitution because its promising<br />

pharmacology was extensively characterized<br />

in the literature, although it possessed poor<br />

pharmacokinetic profiles in preclinical<br />

species and was never progressed into clinical<br />

development. L-838417 was shown to have<br />

desirable GABAA subtype selectivity in vitro:<br />

no agonist activity at the alpha-1 subtype but<br />

partial agonism activities at the alpha-2, 69<br />

SPECIALTY PHARMA JULY/AUGUST 2012 Vol 12 No 6


SPECIALTY PHARMA JULY/AUGUST 2012 Vol 12 No 6<br />

70<br />

alpha-3, and alpha-5 subtypes. This in vitro<br />

subtype profile for L-838417 positively<br />

translated to its in vivo pharmacology<br />

profile, with demonstrated preclinical<br />

efficacy in anxiety, muscle relaxation,<br />

inflammatory pain, and neuropathic pain,<br />

and significantly reduced sedation and<br />

ataxia liabilities in rodents and primates.<br />

Deuterated CTP-354 demonstrated<br />

substantial pharmacokinetic improvement<br />

compared to L-838417 in rats and dogs, and<br />

has recapitulated beneficial L-838417<br />

pharmacology in both in vitro and in vivo<br />

studies. The Chung model, a sciatic nerve<br />

ligation model of neuropathic pain, was<br />

performed in the rat to compare oral doses<br />

of CTP-354 first to L-838417, and then to<br />

the standard of care, gabapentin. CTP-354<br />

was efficacious in both studies with a<br />

prolonged pharmacodynamic effect versus<br />

L-838417, which correlated with the higher<br />

CTP-354 plasma level observed at the final<br />

timepoint. In the comparison of CTP-354 to<br />

gabapentin, CTP-354 demonstrated an<br />

excellent dose response and showed<br />

equivalent efficacy to gabapentin at similar<br />

doses. At the doses used in these studies,<br />

CTP-354 was well tolerated in a rat rotarod<br />

study assessing potential for sedation and<br />

ataxia. The excellent pharmacokinetic and<br />

pharmacology profiles of CTP-354 support<br />

the potential use as a non-sedating treatment<br />

of spasticity and neuropathic pain.<br />

C-21359: A Deuterium-<br />

Stabilized Enantiomer<br />

of Lenalidomide<br />

Concert has developed C-21359 as a<br />

novel deuterated enantiomer of racemic<br />

lenalidomide (Revlimid ® ), an oncology<br />

agent indicated for use in multiple myeloma<br />

and in myelodysplastic syndromes (MDS).<br />

Revlimid contains a mixture of R and S<br />

enantiomers that interconvert in vivo. While<br />

the S enantiomer appears to possess the<br />

beneficial activities associated with<br />

Revlimid, dosing of the single S enantiomer<br />

is not considered useful, as it would rapidly<br />

convert in vivo to the R/S mixture. Concert<br />

has discovered a specific deuterium<br />

modification pattern that optimally slows<br />

the interconversion of enantiomers, thus<br />

enabling administration of the more<br />

beneficial S enantiomer with minimal<br />

exposure to the R enantiomer.<br />

C-21359 demonstrates enhanced<br />

activity versus Revlimid in<br />

immunomodulatory and anti-cancer in vitro<br />

efficacy assays. Deuterium-stabilized, single<br />

S-enantiomer dosing drives this<br />

improvement in pharmacological efficacy,<br />

as deuterated racemic lenalidomide<br />

derivatives show no improvement over<br />

Revlimid in these assays. C-21359 is<br />

expected to be useful for the treatment of<br />

multiple forms of cancer and<br />

myelodysplastic syndrome, with the<br />

potential to expand into non-cancer<br />

indications.<br />

Summary<br />

The aforementioned examples show<br />

that deuterium modification may be used<br />

broadly to improve upon previously known<br />

compounds or their analogs, in turn offering<br />

potential benefits in a wide range of<br />

therapeutic areas. It should be noted,<br />

however, that deuterium effects on the<br />

overall metabolism of a drug are not<br />

predictable a priori. For example, deuterium<br />

substitution may have no observable effect,<br />

or may cause metabolism to shift<br />

preferentially to another existing pathway. In<br />

select cases, Concert’s deuterium<br />

modifications can impart significant<br />

improvements to the metabolic properties of<br />

a drug. Thus far, this relatively new<br />

approach to medicinal chemistry has yielded<br />

a number of promising drug candidates with<br />

differentiated therapeutic properties. n<br />

Dr. Philip<br />

Graham<br />

Vice President of<br />

Program <strong>Development</strong>,<br />

Concert<br />

Pharmaceuticals<br />

Dr. Philip Graham currently serves as Vice President of<br />

Program <strong>Development</strong> at Concert Pharmaceuticals. He<br />

has more than 20 years experience in the pharmaceutical<br />

and biotechnology industry. Prior to joining Concert, Dr.<br />

Graham was Vice President of Product Management and<br />

Imaging at EPIX Pharmaceuticals, where he also had<br />

management responsibility for chemical and analytical<br />

development along with pharmacology and toxicology.<br />

Before joining EPIX, Dr. Graham worked at Eli Lilly and Co.<br />

for more than 5 years in analytical development. He earned<br />

his BS with honors from the University of Otago, New<br />

Zealand, and his PhD in Analytical Chemistry from the<br />

University of Massachusetts, Amherst.<br />

Dr. Julie Fields<br />

Liu<br />

Director, Research<br />

Management,<br />

Concert<br />

Pharmaceuticals<br />

Dr. Julie Fields Liu is currently Director, Research<br />

Management at Concert Pharmaceuticals, where she<br />

serves as a Program Team Leader, Intellectual<br />

Property/Research Liaison, and Manager of a multi-year<br />

outsourced chemistry collaboration. She is a medicinal<br />

chemist and has 12 years of experience in the<br />

pharmaceutical and biotechnology fields. Prior to joining<br />

Concert as one of the first 10 employees, Dr. Liu worked<br />

at Millennium Pharmaceuticals in the areas of oncology,<br />

inflammation, and metabolic disease. She earned her BS<br />

in Chemistry with honors and distinction from the<br />

University of North Carolina at Chapel Hill, and her PhD in<br />

Organic Chemistry from the University of California,<br />

Berkeley.<br />

David Turnquist<br />

Director of Analytical<br />

Chemistry,<br />

Concert<br />

Pharmaceuticals<br />

David Turnquist currently serves as Director of Analytical<br />

Chemistry at Concert Pharmaceuticals. He has more than<br />

14 years of experience in the pharmaceutical industry.<br />

Prior to joining Concert, Mr. Turnquist worked at Vertex<br />

Pharmaceuticals and Merck Research Laboratories serving<br />

in a variety of disciplines from Physical and Analytical<br />

Chemistry to Analytical <strong>Development</strong> to CMC Project<br />

Management. He earned his BS in Biology and Chemistry<br />

from the University of North Carolina at Chapel Hill and his<br />

MBA from Boston College.


Executive<br />

Summary<br />

Q: Can you please discuss AD and its current<br />

prevalence in the US?<br />

A: As the number of people 65 years and older in the US continues to<br />

grow, the number of AD patients will increase. Alzheimer’s is the most<br />

common cause of dementia, and 13% of all individuals over the age of<br />

65, or one in eight, have this disease. Alzheimer’s affects the parts of the<br />

brain that control memory, thought, and language. Doctors do not know<br />

the exact cause of this disease; however, medical research is working to<br />

discover what causes it and how to best treat this condition. Alzheimer’s is<br />

progressive and affects people in different ways. Although each case of<br />

Alzheimer’s is unique to each specific patient, there are similarities in the<br />

signs and symptoms that develop as the disease progresses.<br />

Q: Accera, Inc.’s lead product, Axona ® , targets<br />

cerebral hypometabolism. Can you explain<br />

what cerebral hypometabolism is and how<br />

Axona combats it in AD patients?<br />

A: One of the hallmark signs of Alzheimer’s is the significant drop<br />

in the brain’s ability to metabolize glucose, which is the primary<br />

Holger Kunze<br />

CEO<br />

Accera, Inc<br />

Accera, Inc: Discovering Breakthroughs in<br />

Treating Central Nervous System Disorders<br />

Alzheimer’s disease (AD) is the most common cause of dementia. Thirteen percent of all individuals over the age of<br />

65, or one in eight, have AD. Someone in America develops AD every 69 seconds, and while medications for<br />

treating AD exist, there is no cure. Accera, Inc., a privately held, fully integrated development and commercialization-<br />

stage company, is focused on the discovery and development of pioneering therapeutics for serious diseases, such as AD.<br />

Accera’s lead product, Axona ® , currently marketed in the US as a prescription-only medical food for the clinical dietary<br />

management of mild-to-moderate AD, addresses metabolic deficiencies and provides an alternative energy source for the<br />

brains of AD patients. Specialty Pharma recently interviewed Holger Kunze, CEO of Accera, to discuss the company's<br />

novel approach to treating AD by addressing cerebral hypometabolism.<br />

source of fuel for the brain. This is known as cerebral<br />

hypometabolism. Research has shown that decreases in glucose<br />

metabolism can be seen in the brain 10 to 20 years before any<br />

clinical AD symptoms appear. Normal, healthy brains depend on<br />

circulating glucose to carry out most of its functions, including<br />

memories. Decreases in glucose metabolism correlate with decreases<br />

in brain function in AD patients.<br />

Axona is designed to combat cerebral hypometabolism by<br />

providing the brain with an alternative source of fuel. Axona contains<br />

caprylic triglyceride, a medium chain triglyceride that is broken<br />

down by the liver into ketone bodies, which patients can metabolize<br />

to use as fuel for the brain.<br />

Q: How does Axona differ from other currently<br />

available AD treatments?<br />

A: The first class of FDA-approved medications for AD has been<br />

formulated to increase the levels of Acetylcholine, a chemical<br />

responsible for memory in the brain. This class is called<br />

cholinesterase inhibitors. The primary members of this class are<br />

Exelon and Aricept. While these treatments have been proven<br />

SPECIALTY PHARMA JULY/AUGUST 2012 Vol 12 No 6<br />

71


SPECIALTY PHARMA JULY/AUGUST 2012 Vol 12 No 6<br />

72<br />

effective, many AD specialists believe patients benefit from a<br />

conjunctive or complementary plan that combines different courses<br />

of treatment.<br />

In addition, cholinesterase inhibitors do not alleviate the<br />

cerebral hypometabolism present in the brains of these patients.<br />

Axona’s ability to provide the brain with an alternative source of<br />

energy makes it different from all other currently available AD<br />

treatments.<br />

Q: Axona comes in a powder form. Can you<br />

explain how Axona should be administered?<br />

A: Axona is available in individual powder packets of 40 g per<br />

packet. It is recommended patients start with a reduced dose of 10 g<br />

a day and gradually work their way up to a full dose over the course<br />

of a week. Once acclimated to Axona, patients take only one packet<br />

of Axona once a day, shortly after breakfast. Axona should be added<br />

to 4 to 8 ounces of water or other liquids and be fully blended until<br />

mixed. These are the standard, recommended dosing instructions for<br />

Axona; however, each patient should work with his or her physician<br />

to determine the best and most effective dosing strategy.<br />

Q: Axona is classified as a medical food. What is<br />

a medical food, and how is it regulated?<br />

A: The category of medical foods was defined by the FDA in the<br />

Orphan <strong>Drug</strong> Act in 1988 as a food that is formulated to be<br />

consumed orally under the supervision of a physician, and which is<br />

intended for the specific, dietary management of a disease or<br />

condition. Medical foods are not subject to the FDA’s full approval<br />

process for medication; however, medical foods have a generally<br />

regarded as safe (GRAS) status under the FDA. Medical foods are<br />

more potent and effective than nutraceuticals and supplements.<br />

Supplements are intended for normal, healthy adults and do not<br />

require any pre-market efficacy testing, while medical foods are<br />

intended to treat patients with a specific illness or disease. Medical<br />

foods, unlike nutraceuticals and supplements, MUST be taken under<br />

medical supervision.<br />

Q: Is there clinical data showing that Axona is<br />

safe and effective?<br />

A: Axona has been tested in both a single-dose study and clinical<br />

trials with patients with mild-to-moderate AD, as well as healthy<br />

elderly volunteers. The single-dose study was conducted on 20<br />

patients diagnosed with AD or mild cognitive impairment. The<br />

results showed a positive response, with improvement in paragraph<br />

recall and the AD Disease Assessment Scale - Cognitive subscale<br />

(ADAS-Cog) test. Peer-reviewed results were published in<br />

Neurobiology of Aging, in 2004.<br />

Axona was also studied in a double-blind, randomized,<br />

placebo-controlled study performed at multiple US sites. The trials<br />

involved 152 patients with mild-to-moderate AD over a period of 90<br />

days. As Axona is designed to function as a complementary therapy,<br />

about 80% of those patients were receiving standard-of-care AD<br />

therapy, including the use of cholinesterase inhibitors and NMDA<br />

anatagonists. The results showed a significant improvement in<br />

cognitive function in patients taking Axona after 45 days and<br />

demonstrated a decline in patients in the placebo group. The results<br />

also showed that cognitive benefits could be maintained over a<br />

period of time in the Axona patients.<br />

Peer-reviewed results were published in Nutrition &<br />

Metabolism in August 2009 in the article titled Study of the<br />

Ketogenic Agent AC-1202 in Mild-to-Moderate Alzheimer's<br />

Disease: a Randomized, Double-Blind, Placebo-Controlled,<br />

Multicenter Trial.<br />

Q: Can Axona be taken with the cholerestinase<br />

inhibitors that are currently available for AD<br />

patients?<br />

A: Axona has been designed to complement the existing AD<br />

therapies currently on the market. Patients in clinical trials were<br />

permitted to take commonly prescribed AD medications, and<br />

additional improvements following Axona administration were<br />

observed even for those patients taking these other medications.<br />

Q: Who do you see as Axona’s chief competitors?<br />

A: Among currently available, prescription-only therapies for AD,<br />

Axona has no direct competitors, as no other AD medication targets<br />

cerebral hypometabolism. More importantly, as Axona can be taken<br />

along with cholinesterase inhibitors, it is not competing directly<br />

with this class.<br />

Q: Does Accera currently have any partnerships?<br />

A: Accera is not currently affiliated with any partners, but is<br />

actively seeking strategic partners interested in advancing<br />

ccera’s technology to other indications and promoting global<br />

adoption of Axona. Additionally, Accera is exploring strategic<br />

partnerships to maximize the commercial success of Axona in<br />

and outside the US. n


3M<br />

Company Pg Phone Web Site<br />

Aveva DDS<br />

BASF<br />

Bend Research<br />

Catalent<br />

DDL23<br />

Early <strong>Drug</strong> <strong>Development</strong> Summit 2012<br />

Innercap Technologies<br />

LifeSciencePR<br />

Ligand Pharmaceuticals<br />

Novozymes<br />

Nusil Technology<br />

Particle Sciences<br />

PharmaCircle<br />

ResearchDx<br />

Universe of Pre-filled Syringes and Injection Devices<br />

UPM Pharmaceuticals<br />

Xcelience<br />

5<br />

7<br />

16<br />

75<br />

76<br />

47<br />

58<br />

37<br />

65<br />

9<br />

3<br />

13<br />

Insert<br />

31<br />

2<br />

68<br />

17<br />

11<br />

800-643-8086<br />

954-624-1374<br />

800-706-8655<br />

877-576-8457<br />

+44(0) 1275 849019<br />

+44 (0) 20 7202 7690<br />

813-837-0796<br />

800 - 724-2372<br />

610-861-4701<br />

760-436-1199<br />

866-225-9195<br />

608-643-4444<br />

www.3m.com/MTS<br />

www.avevaDDS.com<br />

www.soluplus.com<br />

www.bendresearch.com<br />

www.catalent.com<br />

www.ddl-conference.org.uk<br />

www.eddsummit.com<br />

www.innercap.com<br />

www.lifesciencepr.net<br />

www.captisol.com<br />

www.stabilizeyourformulation.com<br />

www.nusil.com/ddd<br />

www.particlesciences.com<br />

www.Pharmacircle.com<br />

www.researchdx.com<br />

www.pda.org/prefilled2012<br />

www.upm-inc.com<br />

www.xcelience.com<br />

<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

73


<strong>Drug</strong> <strong>Development</strong> & <strong>Delivery</strong> July/August 2012 Vol 12 No 6<br />

74<br />

On Behalf of Private Equity<br />

By: John A. Bermingham<br />

*Disclaimer: This is not a political article to support a candidate for office.<br />

Throughout the past decade, I have worked for four different<br />

private equity firms as the CEO of one of their portfolio<br />

companies. I, like you, have been listening to various parties<br />

drag private equity firms through the mud disseminating inaccurate<br />

information about what private equity firms do to companies and their<br />

people. I have been there, so I believe I am qualified to offer a first-<br />

hand opinion on private equity firms.<br />

Private equity firms often buy distressed companies with the<br />

intention of turning them around, building them up, and then selling<br />

them in 5 to 7 years for a profit. Most often, this means most of the<br />

people working in the company keep their jobs following the<br />

acquisition of the company by a private equity firm and, when the<br />

company is sold in later years, the people continue on with the<br />

company and its new owners. However, this is not always the case as<br />

you will see further on.<br />

Private equity firms do not buy companies in order to fire all of<br />

the people and shut down the business. Because many of the<br />

companies acquired by private equity firms are in serious trouble, one<br />

of the most common problems in a distressed company is there are<br />

too many people in the company. This is due to the CEO not wanting<br />

to lay off people. I found this to be the case in all four private equity-<br />

owned companies I turned around.<br />

As an example, let’s say that you become the CEO of a distressed<br />

company that does $50 million per year in revenue and is losing $5<br />

million per year. Five years earlier, the company had annual revenue<br />

of $100 million and required a headcount of 500 people. The<br />

company still has 500 employees with an overhead expense of $12<br />

million per year, and your analysis of the head count in the company<br />

indicates you only need 250 people to generate $50 million in<br />

revenue.<br />

Simplistic financial theory shows that reducing headcount by<br />

50% should cut your overhead expense by 50%, so your expense<br />

reduction for people would be $6 million. If you are losing $5 million<br />

per year and cut expenses $6 million per year, you should generate $1<br />

million in profit, assuming everything else remains the same.<br />

The idea here is that by reducing headcount to 250 people, you<br />

save the jobs of the 250 people who are left. If you kept everybody on<br />

board in the company, under current conditions, the company would<br />

eventually go bankrupt and then 500 people would lose their jobs!<br />

So when you hear that such and such company was acquired by a<br />

private equity firm and they fired lots of people, you most likely have<br />

only part of the story. The same holds true when you hear of a private<br />

equity firm buying a company and then shut it down and everyone<br />

lost their job. The private equity firm most likely acquired a distressed<br />

company and had to shut it down because they could not save it.<br />

This is not to say that all private equity firms are good. I have<br />

seen instances in which a private equity firm acquires a company and<br />

brings in a new CEO and management team to turn around and<br />

stabilize the company. Once accomplished, they put the company up<br />

for sale and in many cases, sell the company to a strategic buyer<br />

(another company that is in the same business or industry). The<br />

private equity firm normally makes a lot of money on this type of<br />

transaction.<br />

The acquiring company will look to absorb the business into its<br />

company with a minimal increase in headcount. Hence, the acquiring<br />

company has no need for most of the people in the company it just<br />

acquired, so all of those people lose their jobs.<br />

There is good and bad in almost everything, including private<br />

equity firms. I just wanted to let you know that not all private equity<br />

firms are bad, even though some of the media (as well as some<br />

politicians) portray them as evil personified. u<br />

B I O G R A P H Y<br />

John A. Bermingham is currently the Co-President<br />

and COO of AgraTech, a biotech enterprise focused on<br />

chitosan, a biomaterial processed from crustacean<br />

shells (shrimp, crawfish, crab, etc). He was the<br />

President & CEO of Cord Crafts, LLC, a leading<br />

manufacturer and marketer of permanent botanicals.<br />

Prior to Cord Crafts, he was President & CEO of Alco<br />

Consumer Products, Inc., an importer of house ware,<br />

home goods, pet, and safety products under the Alco<br />

brand name and through licenses from the ASPCA and Red Cross. He successfully<br />

turned around the company in 60 days and sold Alco to a strategic buyer. Mr.<br />

Bermingham was previously the President & CEO of Lang Holdings, Inc. (an<br />

innovative leader in the social sentiment and home décor industries) and<br />

President, Chairman, and CEO of Ampad (a leading manufacturer and distributor<br />

of office products). With more than 20 years of turnaround experience, he also<br />

held the positions of Chairman, President, and CEO of Centis, Inc., Smith Corona<br />

Corporation, and Rolodex Corporation. He turned around several business units of<br />

AT&T Consumer Products Group and served as the EVP of the Electronics Group<br />

and President of the Magnetic Products Group, Sony Corporation of America. Mr.<br />

Bermingham served 3 years in the U.S. Army Signal Corps with responsibility for<br />

Top Secret Cryptographic Codes and Top Secret Nuclear Release Codes, earned his<br />

BA in Business Administration from Saint Leo University, and completed the<br />

Harvard University Graduate School of Business Advanced Management Program.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!